to my parents for their unyielding support and encouragement

126
1 EVALUATION OF BACILLUS SUBTILIS R0179 ON GENERAL WELLNESS, GASTROINTESTINAL SYMPTOMS AND GASTROINTESTINAL VIABILITY: A RANDOMIZED, DOUBLE-BLIND, PLACEBO-CONTROLLED, CLINICAL TRIAL IN HEALTHY ADULTS By ABDULAH HANIFI A THESIS PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF MASTER OF SCIENCE UNIVERSITY OF FLORIDA 2013

Upload: others

Post on 21-Jan-2022

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: To my parents for their unyielding support and encouragement

1

EVALUATION OF BACILLUS SUBTILIS R0179 ON GENERAL WELLNESS, GASTROINTESTINAL SYMPTOMS AND GASTROINTESTINAL VIABILITY: A

RANDOMIZED, DOUBLE-BLIND, PLACEBO-CONTROLLED, CLINICAL TRIAL IN HEALTHY ADULTS

By

ABDULAH HANIFI

A THESIS PRESENTED TO THE GRADUATE SCHOOL

OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF

MASTER OF SCIENCE

UNIVERSITY OF FLORIDA

2013

Page 2: To my parents for their unyielding support and encouragement

2

© 2013 Abdulah Hanifi

Page 3: To my parents for their unyielding support and encouragement

3

To my parents for their unyielding support and encouragement

Page 4: To my parents for their unyielding support and encouragement

4

ACKNOWLEDGMENTS

I would like to thank my advisor and mentor Dr. Wendy Dahl whose knowledge

and guidance has invariably led to my success in a field of science that I have now

grown to love. Thank you Dr. Dahl for taking a chance on me at a time when I was lost

in a sea of opportunity. I would also like to extend the most sincere gratitude to my

colleagues: Dr. Tyler Culpepper for his support and guidance in the field of

microbiology, Amanda Ford for her invaluable aid, advice and willingness to help me

learn what it takes to run a clinical trial, Lauren Khouri for her continued patience and

willingness to help, Allyson Radford for her expertise and guidance in regulatory affairs

and Dr. Younis Salmean for his sage wisdom in navigating the currents of graduate

studies. I would also like to thank the study volunteers who dedicated some of their

valuable time towards helping me achieve my graduate degree.

Page 5: To my parents for their unyielding support and encouragement

5

TABLE OF CONTENTS Page

ACKNOWLEDGMENTS .................................................................................................. 4

LIST OF TABLES ............................................................................................................ 8

LIST OF FIGURES .......................................................................................................... 9

LIST OF ABBREVIATIONS ........................................................................................... 10

ABSTRACT ................................................................................................................... 11

CHAPTER

1 LITERATURE REVIEW .......................................................................................... 13

Introduction to the Intestinal Microbiome ................................................................ 13

Origins of the Probiotic Concept ............................................................................. 15 Modern Definition of Probiotics ............................................................................... 16

Identification and Selection of Probiotic Strains ...................................................... 16 Technical Aspects ............................................................................................ 17

Physiological Aspects ....................................................................................... 18 Functional Aspects ........................................................................................... 19

Evaluating the Safety of Probiotic Bacteria ............................................................. 20 Guidelines for Evaluating the Safety of Bacterial Strains.................................. 20

Systemic infections .................................................................................... 21 Deleterious metabolic activity ..................................................................... 21

Gene transfer ............................................................................................. 22 Excessive immune stimulation ................................................................... 22

In Vitro and Animal Studies .............................................................................. 23 Clinical Trials .................................................................................................... 24

Quality of Life Assessment ..................................................................................... 25 Development of the GSRS ............................................................................... 25

Initial Validation of the GSRS ........................................................................... 26 An Updated GSRS ........................................................................................... 27

Internal consistency reliability .................................................................... 28 Construct validity ........................................................................................ 29

Discriminant validity ................................................................................... 30 Responsiveness ......................................................................................... 31

Determination of Healthy Status in Research Participants ...................................... 32 Bacillus subtilis as a Probiotic ................................................................................. 35

In Vitro Studies ....................................................................................................... 41 The Safety of Bacillus subtilis ........................................................................... 41

The Safety of Bacillus subtilis R0179 ............................................................... 42 Animal Studies ........................................................................................................ 43

Germination of Bacillus subtilis in the Gastrointestinal Tract ............................ 43 B. subtilis in the Human Intestinal Tract ........................................................... 46

Page 6: To my parents for their unyielding support and encouragement

6

Potential Health Benefits of Bacillus subtilis ........................................................... 47 The Effect of B. subtilis on Immune Function ................................................... 47

The Effect of B. subtilis on Bone Metabolism ................................................... 49 The Use of B. subtilis as Bowel Preparation for Colonoscopy .......................... 50

Conclusions ............................................................................................................ 51

2 PURPOSE .............................................................................................................. 53

3 METHODS AND PROCEDURES ........................................................................... 54

Study Design .......................................................................................................... 54

Inclusion and Exclusion Criteria before Attaining Consent...................................... 54 Recruitment ............................................................................................................ 55

Baseline .................................................................................................................. 56 Randomization and Intervention ............................................................................. 57

Washout and Post Intervention ............................................................................... 58 Compensation ......................................................................................................... 58

Stool Protocol ......................................................................................................... 58 Statistical Methods .................................................................................................. 59

Equivalence Testing ......................................................................................... 59 B. subtilis Viability ............................................................................................. 61

4 RESULTS ............................................................................................................... 65

Participants ............................................................................................................. 65

Daily Questionnaire Analysis .................................................................................. 65 Gastrointestinal Symptom Rating Scale Analysis ................................................... 69

Viability of B. subtilis R0179 in Humans.................................................................. 70

5 DISCUSSION AND CONCLUSIONS ...................................................................... 96

Discussion .............................................................................................................. 96 Conclusions .......................................................................................................... 100

APPENDIX

A IRB APPROVAL ................................................................................................... 101

B INFORMED CONSENT ........................................................................................ 102

C GLOBAL PHYSICAL ACTIVITY QUESTIONNNAIRE........................................... 112

D DAILY QUESTIONNAIRE ..................................................................................... 115

E GASTROINTESTINAL SYMPTOM RESPONSE SCALE ..................................... 118

LIST OF REFERENCES ............................................................................................. 120

Page 7: To my parents for their unyielding support and encouragement

7

BIOGRAPHICAL SKETCH .......................................................................................... 126

Page 8: To my parents for their unyielding support and encouragement

8

LIST OF TABLES

Table page 1-1 Syndrome assignments of the SF-36, PGWB, and GSRS ................................. 52

3-1 Daily Questionnaire syndrome assignments....................................................... 63

3-2 Gastrointestinal Symptom Rating Scale syndrome assignments........................ 64

4-1 Participant demographics ................................................................................... 72

4-2 Daily Questionnaire syndrome equivalence testing ............................................ 73

4-3 Daily Questionnaire symptom equivalence testing ............................................. 82

4-4 Gastrointestinal Symptom Rating Scale equivalence testing .............................. 89

4-5 Gastrointestinal viability of Bacillus spores ......................................................... 95

Page 9: To my parents for their unyielding support and encouragement

9

LIST OF FIGURES

Figure page 3-1 Study design ....................................................................................................... 62

4-1 Participant flow diagram ..................................................................................... 71

4-2 Gastrointestinal distress syndrome ..................................................................... 76

4-3 Cephalic syndrome ............................................................................................. 77

4-4 Epidermal syndrome ........................................................................................... 78

4-5 Ear nose and throat syndrome ........................................................................... 79

4-6 Behavioral syndrome .......................................................................................... 80

4-7 Emetic syndrome ................................................................................................ 81

4-8 Constipation symptom ........................................................................................ 84

4-9 Diarrhea symptom .............................................................................................. 85

4-10 Fatigue symptom ................................................................................................ 86

4-11 Bowel movement frequency ............................................................................... 87

4-12 Hours of sleep .................................................................................................... 88

4-13 GSRS abdominal pain syndrome ....................................................................... 90

4-14 GSRS reflux syndrome ....................................................................................... 91

4-15 GSRS indigestion syndrome............................................................................... 92

4-16 GSRS constipation syndrome............................................................................. 93

4-17 GSRS diarrhea syndrome................................................................................... 94

4-18 Gastrointestinal viability ...................................................................................... 95

Page 10: To my parents for their unyielding support and encouragement

10

LIST OF ABBREVIATIONS

CFU Colony Forming Units DQ Daily Questionnaire EFSA European Food Safety Administration

FAO Food and Agricultural Organization

FDA Food and Drug Administration GERD Gastroesphogeal reflux disease GI Gastrointestinal GPAQ Global Physical Activity Questionnaire GRAS Generally recognized as safe GSRS Gastrointestinal Symptom Rating Scale

IBS Irritable Bowel Syndrome

ICC Intraclass coefficients PBS Phosphate buffered saline PGWB Psychological general well-being index QPS Qualified Presumption of Safety

SF-36 Short Form 36

SP Species

WHO World Health Organization

Page 11: To my parents for their unyielding support and encouragement

11

Abstract of Thesis Presented to the Graduate School of the University of Florida in Partial Fulfillment of the

Requirements for the Degree of Master of Science

EVALUATION OF BACILLUS SUBTILIS R0179 ON GENERAL WELLNESS, GASTROINTESTINAL SYMPTOMS AND GASTROINTESTINAL VIABILITY: A

RANDOMIZED, DOUBLE-BLIND, PLACEBO-CONTROLLED, CLINICAL TRIAL IN HEALTHY ADULTS

By

Abdulah Hanifi

December 2013

Chair: Wendy Dahl Major: Food Science and Human Nutrition

Bacillus subtilis R0179 has been marketed in Asia as the probiotic Medilac®.

This probiotic formulation is composed of Enterococcus faecium R0026 and Bacillus

subtilis R0179 in a 9:1 ratio. However, prior research has not evaluated B. subtilis

R0179 as a single probiotic agent at doses higher than 0.1 billion CFU/capsule. To

confirm the oral dose-response tolerance of B. subtilis R0179, a 6-week randomized,

double-blind, placebo-controlled clinical trial in healthy adults (n=81), 18-50 years of

age, was conducted. Methods: Participants were randomized to one of three B. subtilis

R0179 dosages (0.1, 1 and 10 billion cfu/capsule/day) or placebo for a 4-week

intervention period. General wellness was evaluated using a daily questionnaire (DQ)

composed of six syndromes rated on a scale of 0 (none) to 6 (very severe):

gastrointestinal distress, cephalic, ear-nose-throat, behavioral, emetic, and epidermal.

Gastrointestinal symptoms were evaluated pre-intervention, during intervention and post

intervention with the Gastrointestinal Symptom Rating Scale (GSRS). The GSRS

contains five syndromes rated on a scale of 1 (no discomfort) to 7 (very severe

discomfort): abdominal pain, indigestion, reflux, constipation and diarrhea. Three stool

Page 12: To my parents for their unyielding support and encouragement

12

samples were obtained, at pre-intervention, intervention and post intervention time

points to assess the viability of B. subtilis in the human gastrointestinal tract. Symptom

data were analyzed using the equivalence testing method to assess dose response by

week. Kruskall-Wallis tests were used to compare pre-intervention and intervention

syndrome scores for both the GSRS and DQ. Gastrointestinal viability was compared

on a per week by treatment basis using ANOVA and Tukey-Kramer HSD when

appropriate. Results: General wellness and gastrointestinal function was not adversely

affected by oral consumption of B. subtilis R0179 at any dose. Mean intervention DQ

syndromes were not different from baseline and ranged from 0.04 to 0.7. Mean

intervention GSRS syndromes were not different from baseline and ranged from 1.1 to

1.9. Fecal viable counts of B. subtilis R0179 demonstrated a dose response ranging

from 101 to 105 CFU/g. No side effects of any kind were reported. Conclusions: Overall,

the results demonstrate that consumption of B. subtilis R0179 was well tolerated by

healthy human adults and is viable in the human gastrointestinal tract.

Page 13: To my parents for their unyielding support and encouragement

13

CHAPTER 1 LITERATURE REVIEW

Introduction to the Intestinal Microbiome

The human gastrointestinal tract is home to a large number of microbes which

first colonize humans during the fetal stage of development. This colonization process

continues during birth and up through adolescence (1, 2). The composition of these

microbes varies from person to person and is impacted by personal and environmental

factors such as the decision to breast or formula feed an infant (3) and the nosocomial

spread of microbes in maternity wards (4). During late adolescence the intestinal

microbiome begins to stabilize in both number and composition (5). In healthy adults,

the general intestinal microbiome is stable and remains so until old age when a decline

in gastrointestinal function begins to cause a shift in the number and variety of species

(6).

The variation of microbial species in the human gastrointestinal tract differs from

one individual to another but overall, nearly 97% of gut microbes are strict anaerobes

while the remaining 3% are aerobic. These consist of autochthonous species

(permanent residents) and allochothonous species (transitory colonizers) that make up

the 400 to 1000 species present throughout the gastrointestinal tract (7). Autochthonous

species are typically acquired in the early years of human life while the allochothonous

species enter the gastrointestinal tract through activities like ingestion of food.

Allochothonous species temporarily colonize the gastrointestinal tract and are

subsequently excreted along with feces. The identification of these microbes and the

roles they play in healthy gastrointestinal function is consistently being evaluated with

international projects such as the US National Institutes of Health’s Human Microbiome

Page 14: To my parents for their unyielding support and encouragement

14

Project, the European Commission’s Metagenomics of the Human Intestinal Tract

project and the Canadian Microbiome Initiative (8). These projects have provided great

insight into the importance of gastrointestinal microbes and the association of certain

disease states to differing microbiome compositions. However, a conclusive definition of

a healthy human microbiome is not yet defined.

Current research has only just begun to piece together the important components

of a healthy microbiome. A majority of microbiota research has focused on

understanding the composition of the intestinal microbiome in sick or diseased

individuals and then comparing them to controls. The main difference identified between

a healthy and an unhealthy microbiome is the high and low diversity of species,

respectively (9). Recent studies have demonstrated a link between low diversity in the

human intestinal microbiome and both obesity and inflammatory bowel disease (10, 11).

These disease states are a result of the inability of the gastrointestinal bacteria to resist

ecologic stressors like pathogenic bacteria and antibiotic usage (8).

Ecologic stressors cause temporary imbalances of the intestinal microbiome and

can either have a negative impact (dysbiosis) or a positive impact (probiosis) on the

function of the gut. For example, antibiotic usage is regarded as a dysbiotic event

because of its ability to reduce the number and composition of autochthonous

organisms in the gut which increases the chances of pathogenic organisms to take up

residence in the gut (12). Disease states like antibiotic-associated diarrhea, metabolic

syndrome and atopic eczema have been associated with dysbiosis of the intestinal

microbiome (13). The converse is true for probiosis of the intestinal microbiome which

may serve to maintain or reestablish the homeostasis between host and microbiome.

Page 15: To my parents for their unyielding support and encouragement

15

The current state of research has begun to enumerate the health benefits provided to

humans by their intestinal microbiome. The purpose of this literature review is first to

identify the origins of the probiotic concept, then to define modern probiotics and outline

their regulation, next to understand how research is conducted on novel probiotic

agents and finally to evaluate the usage of a spore-forming bacteria as a probiotic.

Origins of the Probiotic Concept

Elie Metchnikoff’s Prolongation of Life, published in 1907, is one of the first

substantial works evaluating dysbiosis and probiosis. In this work, he examines how

various mechanisms that modulate the intestinal microbiome can prolong life (14). In a

section titled “Intestinal Putrefaction Shortens Life”, he examines the role of bacteria in

the bowels. He begins to describe that in the first hours after birth the microbes present

in the infant’s intestinal tract thrive on shed intestinal mucosal cells. The check against

the overgrowth of these opportunistic microbes is the consumption of mother’s milk

which promotes the colonization of the infantile gastrointestinal tract with Bifidobacteria

(14). Metchnikoff then diverts his attention to the fact that because microbes cause the

natural putrefaction of food outside of the bowels they may also be the cause of the

putrefaction of the bowels. Some foods like milk, however, resist putrefaction, because

of the preservative act of souring (14). He attributes this preservation of food to the

production of lactic acid, a by-product of glucose fermentation.

Metchnikoff continues this train of thought by reviewing research that evaluated

the anti-putrefactive properties of the consumption of yoghurt derived Lactobacilli

cultures and lactic acid (14). Metchnikoff cites numerous studies where consumption of

lactic acid producing microbes decreased gastrointestinal symptoms and increased

quality of life. The research conducted was anecdotal by today’s standards but the

Page 16: To my parents for their unyielding support and encouragement

16

conclusions that Metchnikoff draws from them are very important to history of probiotic

usage. He advocates for the consumption of live microbes and lactic acid that suppress

the naturally putrefactive intestinal microbes much in the same way that a mother’s

breast milk suppresses the overgrowth of microbes in an infant (14). These concepts of

probiosis and dysbiosis have since evolved and are continually being revised based on

scientific discoveries.

Modern Definition of Probiotics

The concept of probiosis and the probiotic effect of microbes on the

gastrointestinal tract is a heavily researched topic throughout the world. In the past 20

years, the number of research publications on intestinal microbiota has increased

fivefold with about 500 publications occurring in 2009 and almost double that occurring

in 2012 (15). The importance of this field of research has reached a global scale and so

the Food and Agriculture Organization (FAO) and the World Health organization (WHO),

have published a working group report on probiotics and their importance (16). They

define probiotics as “live microorganisms which when administered in adequate

amounts confer a health benefit on the host” (16). The identification of these

microorganisms is the first step in a process to determine the safety and efficacy of

probiotics. The report continues on to describe preferred identification and evaluation

methods of these microorganisms.

Identification and Selection of Probiotic Strains

The process of selecting a particular type of bacteria for use as a probiotic and

evaluating it for safety begins with its identification (17). The first step in identifying a

microbe is by characterizing its genus, species, strain and origin. Identifying the

bacterium in this manner also allows scientists to assess its pathogenicity by comparing

Page 17: To my parents for their unyielding support and encouragement

17

it to genetically similar microorganisms (18). For example, if a certain species of

pathogenic bacteria typically produces enterotoxins (proteins produced by bacteria

which can cause gastrointestinal disorders like vomiting and diarrhea), and is antibiotic

resistant, then this organism would be unfit for human consumption. Similarly, if the

bacteria being identified is genetically similar to the pathogenic bacteria then it may be

ruled out as a probiotic. The gold standard by which a bacterium can be identified at the

strain specific level is through DNA-DNA hybridization (16). This technique allows the

DNA of several different species of bacteria to be compared against one another. The

identification process also includes a characterization of the bacteria’s technical,

physiological and functional aspects. These aspects act as a set of criteria to assess the

probiotic activity of the bacteria in vitro using models of the human gastrointestinal tract

(18).

Technical Aspects

The technical aspects of bacteria that are characterized are growth properties

during processing, and viability during transport and storage (18). Evaluating growth

properties in vitro allows scientists to understand the ideal growth conditions of the

bacteria while characterizing the viability of the bacteria during transport and storage

contributes to their mass marketing potential (18). If a bacteria grows readily but is

unable to survive transport and storage then its ability to cause a probiotic effect after

consumption is lost. A third technical aspect which is important to consumers are the

sensory properties of the probiotic (19). If current trends continue, more and more

probiotics will be added to foods forming “functional foods”, which provide a benefit to

the consumer beyond basic sustenance. Evaluation of the sensory properties of a

probiotic in food will contribute to its overall acceptability by consumers.

Page 18: To my parents for their unyielding support and encouragement

18

Physiological Aspects

Physiological aspects of bacteria that are characterized are the ability to

withstand environmental stressors, and the ability of the bacteria to adhere to human

intestinal epithelial cells (18). Environmental stressors can include a lack of nutrient

availability, fluctuations in temperature, and competition among species. Upon

consumption, bacteria must be able to survive the antimicrobial properties of the human

gastrointestinal tract such as stomach acid, bile acid, and pancreatic juices (18).

The ability of the bacteria to survive the acidic conditions of the stomach and

upper gastrointestinal tract are essential for it to begin transitory colonization in the

jejunum and ileum where the majority of bacteria reside. While some colonization does

occur in the stomach and duodenum (101 and 103 respectively), a large number of

bacteria spend a part of the life cycle in the jejunum, ileum and colon (104, 107, and 1012

respectively) (15). The bacteria may also be able to digest bile salts which act as

antimicrobials in the gut, though not all probiotics possess this ability (20). Lastly, the

probiotic bacteria must withstand the various digestive enzymes found in pancreatic

juices. After being exposed to all of these environmental stressors, the bacteria must be

able to adhere to intestinal epithelial cells.

Bacterial adhesion to intestinal epithelial cells allows them to begin transitory

colonization of the lower small intestine and colon (21). The adhesion also serves to

competitively exclude other species from colonization thus increasing the chances of

survival of the bacteria (15). Probiotic bacteria that can withstand the harsh

antimicrobial conditions of the gastrointestinal tract behave just like hardy pathogenic

bacteria, and because they are able to survive, they can competitively exclude the

pathogenic bacteria from adhering to intestinal epithelial cells. This prevents dysbiosis

Page 19: To my parents for their unyielding support and encouragement

19

from occurring. Once it has been demonstrated that a bacteria possesses the

necessary technical and physiological aspects, the functional and potential health

benefits of the bacteria are characterized (18).

Functional Aspects

Primary functional aspects of probiotics include the production of antimicrobials

and stimulation of autochthonous microbes. When considered in tandem, these aspects

serve to divest the gastrointestinal tract of pathogenic bacteria and subsequently

enhance the growth of resident gut microbes (15). The potential health benefits of

probiotic bacteria has been well documented in a myriad of studies and reviews of the

literature (22). Mercenier et al. list some potential health benefits attributed to lactic acid

bacteria which contains 39 distinct items ranging from specific immunomodulatory

properties such as an increase in immunoglobulin A (a gastrointestinal antibody)

production to a general effect of improving well-being (22). Despite the progress being

made identifying the health benefits of different probiotic strains, the knowledge of the

underlying mechanisms are not fully understood (18).

The process of selecting a probiotic strain begins with the identification of that

bacterium on the genus, species, and strain levels. If the bacterium does not possess

pathogenic characteristics then it should be further characterized at the technical,

physiological and functional levels. A potential probiotic strain of bacteria should meet

the following criteria in order to be a good candidate for use as a probiotic: the strain

must grow readily and be viable during transport and storage, the strain must be able to

resist the antimicrobial properties of the human gastrointestinal tract and adhere to

intestinal epithelial cells, the strain must also benefit the host either by competitively

excluding pathogenic bacteria or enhancing the growth of autochthonous microbes.

Page 20: To my parents for their unyielding support and encouragement

20

Evaluating the Safety of Probiotic Bacteria

Regulatory authorities in Europe and North America aim to protect consumers

from pathogenic bacteria being marketed as probiotics. The European Food Safety

Authority (EFSA) recommends that new biologic products pass through a process by

which they receive a qualified presumption of safety (QPS) (18). This process involves

defining the taxonomic unit of the biologic product, assessing the body of knowledge on

the product, assessing its pathogenicity and evaluating its intended use. The American

counterpart to the EFSA is the Food and Drug Administration (FDA). The FDA regulates

probiotic agents as dietary supplements and so they place the burden of proof of safety

on the manufacturer (23). The FDA does not specify any criteria to prove safety, but

rather they note that scientific consensus must demonstrate safety or the manufacturer

must demonstrate safe historical usage of a probiotic agent. For the latter case, the

probiotic bacteria would be generally recognized as safe (GRAS) and would be

approved for sale in food products or as a supplement (18).

Guidelines for Evaluating the Safety of Bacterial Strains

The FAO/WHO working group report cites four possible types of side effects that

may occur from the consumption of bacteria marketed as probiotics. These side effects

were originally identified by Marteau in 2001 and serve as guidelines for evaluating the

safety of a bacterial strain before clinical trials (24). The side effects are:

Systemic Infections

Deleterious metabolic activities

Gene transfer

The excessive stimulation of the host’s immune response.

Page 21: To my parents for their unyielding support and encouragement

21

Systemic infections

Documented occurrences of systemic infections from probiotic consumption are

few in number and have typically occurred in persons with existing medical conditions

(24). For example, four cases of Saccharomyces boulardii fungemia (fungus present in

host blood) were reported in hospitalized patients following S. boulardii therapy (25). S.

boulardii therapy is used to treat diarrhea in enterally fed and Clostridium difficile (a

pathogenic bacteria) infected patients (26).The investigators of this study cited that the

possible routes of infection could have occurred from patient or health provider contact

with intravenous catheters following the handling of S. boulardii packets. One of these

cases resulted in septic shock. The investigators recommended that S. boulardii

packets be handled outside of patient rooms and only with gloves to minimize their

inadvertent spread around susceptible patients (25). In two back to back ecologic

studies located in southern Finland, Saxelin et al. evaluated the prevalence of

bacteremia due to Lactobacilli consumption in 9229 probiotic consumers. This sample

population only produced 20 cases of bacteremia and no isolates of bacteria present in

the blood matched up with any probiotic strains being consumed (27). A follow up study

determined that the average incidence of bacteremia is 0.3 cases/100,000

inhabitants/year in Finland. Investigators concluded that probiotic use did not increase

the incidence of bacteremia (28).

Deleterious metabolic activity

Bacteria affect the metabolism of the small intestine via probiotic and dysbiotic

mechanisms. Some dysbiotic activities that negatively impact metabolic activity include

excessive deconjugation and dehydroxylation of bile salts and excessive degradation of

the intestinal epithelium’s mucosal layer (24). Listeria monocytogenes, a pathogenic

Page 22: To my parents for their unyielding support and encouragement

22

bacterium, deconjugate bile salts, this increases the bacteria’s growth rate and ability to

survive in the human gastrointestinal tract (27, 29). Deconjugation prevents the

precipitation of bile salts in the feces which results in the release of reactive nitrogen

species that accumulate in the colon and contribute to carcinogenesis (30).

Gene transfer

Gene transfer in bacteria occurs when a small molecule of DNA called a plasmid

is passed from one bacterial cell to another. Plasmids are a concern to scientists

because of their ability to spread pathogenic activity among bacteria. For example, if an

enterotoxin-producing strain of bacteria that possesses a plasmid is consumed, then it

is possible for that enterotoxin gene to be passed on to other gut microbes which would

otherwise be harmless (24). This is especially true for plasmids that transfer antibiotic

resistance, especially resistance to the antibiotic vancomycin. Vancomycin is used to

treat infections due to gram positive bacteria like C. difficile (31). These infections

typically occur after methicillin resistant staphylococci nosocomial infections and often

result in mortality because of the bacteria’s ability to survive antibiotic treatment (32).

Excessive immune stimulation

An excess of immunostimulation, can have deleterious effects on the overall

health of an individual (33). The body responds to physiological stressors like infection

or tissue injury through proinflammatory signals which are typically short lived (34). An

excess of this proinflammatory signaling can lead to chronic inflammation and disease

progression (34). Polysaccharides from the cell walls of bacteria typically elicit acute

responses from the host but the continued consumption of a probiotic that elicits this

response can lead to excess immune stimulation. Acute toxicity studies conducted in

murine models test for this excess immune stimulation and evaluate the effects of

Page 23: To my parents for their unyielding support and encouragement

23

consuming high single dosages of a bacteria. A probiotic should not cause dysbiosis

even when consumed at excessively high dosages (15, 35).

Helicobacter pylori is an example of a bacteria that induces excessive immune

stimulation (36). These bacteria invade human intestines and release reactive nitrogen

species which cause oxidative damage to intestinal epithelial cells’ DNA (36). Once the

DNA has been extensively damaged, the cells undergo apoptosis and are shed in the

feces. This elicits an immune response in an effort to rid the intestines of H. pylori.

Chronic H. pylori infection results in ulcers and is a risk factor for carcinogenesis due to

the accumulation of DNA damage in intestinal organs. Furthermore, chronic infection by

H. pylori acts as a compounding factor for further immunostimulation (37). This type of

excessive immunostimulation is one reason why probiotics should be non-invasive,

transitory colonizers of the human gastrointestinal tract.

In Vitro and Animal Studies

In vitro studies typically used to assess the safety of the probiotic bacteria include

genome sequencing of the strain for identification and characterization purposes,

evaluations of antibiotic resistance, gene transfer capability and its ability to withstand

environmental stressors such as gastric juices and stomach acid. Following the

completion of in vitro studies, animal studies are used to assess the pathogenicity,

acute toxicity, and gastrointestinal viability of the bacteria. Animals may also be used to

study probiotic mechanisms or actions that were observed during the in vitro testing

phase (16). If no adverse effects are observed in animal studies and the bacteria has

historically been used safely, then further testing can proceed to clinical trials.

Page 24: To my parents for their unyielding support and encouragement

24

Clinical Trials

Clinical trials involve human volunteers and are divided into Phases I, II, III and

IV (38). Phase I trials are conducted to determine the safety of the biologic agent and

typically consists of less than one hundred volunteers. These trials provide information

on possible symptoms associated with the biologic agent as well as determining a

maximally-tolerated dose (38). Phase II trials are typically conducted on diseased

populations and aim to provide information on the efficacy of the biologic agent to treat

a disease or condition. If improvement of the disease or health condition occurs, in

comparison with a placebo, then scientists will proceed to phase III trials. A phase III

trial expands on the knowledge gained from phase II trials and aims to determine the

advantage of the new biologic agent against standard therapy (38). Phase III trials also

provide information on drug interactions, side effects, and appropriate dosages.

Following the completion of a phase III trial for a new drug, manufacturers can request

to market the drug to the general public and upon approval of that request by a

regulatory agency, they will begin a phase IV trial (38). Phase IV trials are also known

as post-market surveillance studies because rare and slowly developing adverse effects

can more readily be monitored after the drug is made available to the public. Of

important note are recommendations made by the FAO/WHO when evaluating

probiotics in clinical trials. They recommend that during phase II trials (evaluation of

efficacy), validated quality of life assessment tools be used to determine the overall

impact of the probiotic on host health rather than only evaluating its effect on the

disease or health condition (16).

Page 25: To my parents for their unyielding support and encouragement

25

Quality of Life Assessment

Medical treatments for patients presenting with gastrointestinal diseases focus on

the treatment of the disease and at times fail to acknowledge the patients’ general well-

being (39). An evaluation of patients’ symptoms is necessary to assess not only the

reduction in severity of disease but also the overall effect of the intervention on patients’

lives. Gastrointestinal symptoms vary from person to person and often times these

symptoms lack a proper pathophysiologic identification. The subjective nature of

symptom intensity, however, makes it difficult to compare disease severity in different

patients. The development of questionnaire-based instruments to evaluate the presence

and severity of symptoms is necessary to enumerate and compare the overall effect of

conventional medicine across populations. The Gastrointestinal Symptom Rating Scale

(GSRS) is one such instrument. It was developed for the global rating of the severity of

symptoms based on the clinical interview and has been validated in six countries (40,

41).

Development of the GSRS

Svedlund et al. 1988 constructed the original Gastrointestinal Symptom Rating

scale in study participants who presented with irritable bowel syndrome and peptic ulcer

disease. The study evaluated the clinical efficacy of conventional medical therapy for

irritable bowel syndrome (n=101) and peptic ulcer disease (n=103) compared to an

intervention that utilized psychotherapy in conjunction with conventional medical

practices. The symptoms chosen for use in the questionnaire were based off of the

clinical experience of physicians and reports in the literature. The questionnaire

measured symptoms such as epigastric pain, heart burn, increased flatus and nausea;

Page 26: To my parents for their unyielding support and encouragement

26

a full list of symptoms is reported in Table 3-2. The original GSRS utilized four well

defined intensity scores ranging from 0 to 3:

0: No or transient pain

1:Occasional aches and pains interfering with some social activities

2: Prolonged and troublesome aches and pains causing requests for relief and interfering with many social activities

3: Severe or crippling pains with impact on all social activities

Undefined half steps were also used to measure symptoms at higher sensitivities (40):

Initial Validation of the GSRS

The validation of the GSRS was conducted using a subset of participants (n=20)

from the clinical efficacy study conducted by Svedlund et al. The participants were

administered the GSRS in an interview format. Medical records from the past month in

addition to patient responses were utilized to rate the participants with the GSRS. Each

study participant was interviewed and rated twice by two psychiatrists in order to

eliminate interviewer bias. The inter-rater agreement of GSRS scores, determined via

the interview process, was tested for using Cohen’s weighted kappa (Kw) which ranges

in score from 0 to 1. The maximum score of 1 indicates a perfect agreement between

interviewers while a score of 0 indicates no agreement between interviewers (40). A

higher Kw score indicates that the GSRS can be used to accurately gauge the intensity

of the gastrointestinal symptoms it inquires about. All weighted kappa scores measured

were high (ranging 0.86 to 1.00 for individual symptoms and 0.92 to 0.94 for

syndromes) indicating inter-rater agreement was not likely due to chance (40). Despite

high inter-rater agreement, the original GSRS was only utilized to detect the presence

or absence of symptoms. Svedlund et al. did not test the ability of the questionnaire to

Page 27: To my parents for their unyielding support and encouragement

27

detect changes in gastrointestinal symptoms. This severely limits its use in clinical trials

as a diagnostic instrument. Furthermore, Svedlund et al. used an interview process to

administer the questionnaire. In clinical trials, the interview method is unrealistic due to

increased costs and burden on study participants. The investigators concluded that

more studies were necessary in order for the GSRS to be suitable as a diagnostic

instrument in clinical trials.

An Updated GSRS

Revicki et al. 1998 updated the original translations of the GSRS (Svedlund et al.

1988) with simpler terminology replacing words like borborygmus and eructation with

stomach rumbling and burping (42). These changes made the GSRS easier to

understand and increased its appeal for use as a self-administered questionnaire. The

GSRS was validated with respect to the Psychological and General Well-Being scale

(PGWB) and the Medical Outcomes Study short form (SF-36) which evaluate quality of

life. Revicki et al. conducted the validation study using participants (n=516) of a clinical

trial which evaluated Ranitidine as a treatment for gastroesphogeal reflux disease

(GERD). Previous clinical trials utilizing the PGWB and SF-36 in GERD patients

demonstrated the responsiveness of those questionnaires to experimental treatment

(43) (44). Revicki et al. reasoned that if the results generated by the GSRS correlated

well with the PGWB and SF-36 then they could be used together as holistic measures

of experimental treatments rather than only evaluating a reduction in symptoms (39).

The validation process involved evaluating the GSRS’s internal consistency reliability,

construct validity, discriminant validity and responsiveness (42).

Page 28: To my parents for their unyielding support and encouragement

28

Internal consistency reliability

The internal consistency reliability of a questionnaire is a measure of whether or

not the questionnaire means the same thing to different people. Due to the subjective

nature of gastrointestinal symptoms it is necessary for the GSRS to evaluate symptoms

using overarching constructs (42). For example, the GSRS contains a constipation

syndrome which is a mean of three different symptom scores: constipation, feelings of

incomplete evacuation and hard stools. These three symptoms evaluate an overarching

construct, constipation, by asking about some characteristics that are clinically

associated with constipation.

The Cronbach α test statistic is used to measure internal consistency reliability

and ranged from (0.61 to 0.80) for the various syndromes of the GSRS (42). A higher

Cronbach α score (maximum 1, minimum 0) indicates that the symptoms are very

similar in relation to the overarching construct. The Cronbach α score of the constipation

syndrome was 0.80, this is at the upper bound of “good” internal consistency reliability

and indicates that the symptoms are an adequate measure of the syndrome (42).

Next, Revicki et al. used intraclass correlations coefficients (ICC) to measure

how alike the syndromes were between study participants. Higher ICC (maximum 1,

minimum 0) scores indicate that the syndromes meant the same things to different

participants. ICC scores ranged from 0.42 to 0.60. These scores indicate fair to good

agreement between participant’s when taking the self-administered GSRS (42). While

these ICC scores may appear low they provide valuable insight into the validity of the

questionnaire. Symptoms are subjective by nature and so they should inherently mean

different things to different people. An ICC score that is too high would indicate that the

questionnaire does not adequately measure variability and a score that is too low

Page 29: To my parents for their unyielding support and encouragement

29

indicates that the questionnaire allows too much variability in its possible answers. For

example, if a questionnaire asked participants to answer the question, “How old are

you?” the expected ICC score would be very low due to the high variability of possible

answers. In contrast, a perfect score of 1 would indicate that all participants answered

exactly the same to the question. The fact that the internal consistency reliability and

intraclass correlation coefficients of the GSRS falls into the fair to good score range

indicates that it is a true measure of subjective gastrointestinal symptoms. The GSRS is

both sensitive enough to detect differences between study participants and similar

enough to mean the same thing to them (42).

Construct validity

The construct validity of the GSRS is a measure of how well it correlated with the

PGWB and SF-36 quality of life questionnaires. Pearson product moment correlations

were utilized to measure the GSRS’s construct validity (42). Revicki et al., in agreement

with Dimenas et al. and Svedlund et al., hypothesized that GSRS syndrome scores

would negatively correlate with PGWB and SF-36 subscales (Table 1-1). This

hypothesis is based on the idea that gastrointestinal symptoms adversely affect the

general wellness and psychological wellness of individuals (39, 40).

Pearson correlations of the GSRS syndromes and SF-36 sub-scales were all

negative and ranged from -0.12 to -0.44. The reflux syndrome was the only GSRS

syndrome that failed to significantly correlate with the SF-36 in five sub-scales (physical

and emotional role limitations, mental health, and mental component summary and

physical component summary. While the correlations were negative for these five sub-

scales they were not significant (p>0.05). Pearson correlations of the GSRS syndromes

and PGWB sub-scales were all negative and ranged from -0.09 to -0.41. A majority of

Page 30: To my parents for their unyielding support and encouragement

30

the GSRS syndrome and PGWB sub-scale correlations were significant (p<0.0001).

The positive well-being subscale did not significantly correlate with any GSRS

syndrome and the reflux syndrome failed to significantly correlate with depression,

positive well-being and behavioral control PGWB subscales. Revicki et al. conclude that

the GSRS has an acceptable construct validity but they fail to discuss the non-

significant Pearson correlations. Revicki et al. never describe which GSRS values

(baseline or treatment) were used to calculate the Pearson correlations. Considering

that the reflux syndrome correlations were consistently not significant, in a study

population that was receiving Ranitidine therapy to treat GERD, it is likely that the

investigators used treatment GSRS scores to calculate Pearson correlations. This

diminishes the quality of their results because correlation scores could be

underestimated due to Ranitidine’s reduction in symptom severity. For example, the net

change of the reflux syndrome score was -1.23 after 6 weeks of treatment indicating

that PGWB and SF-36 scores would be higher if GSRS scores during the treatment

period would be used. This would reduce the correlation score thus underestimating the

predictive potential of the GSRS syndromes on quality of life. Overall Revicki et al.

conclude that the construct validity of the GSRS is consistent with previous findings (39)

and they support its continued use as an indirect measure of quality of life.

Discriminant validity

Revicki et al. also examined the discriminant validity of the questionnaire which

evaluates the effectiveness of the questionnaire to identify treatment responders from

treatment non-responders (42). Participants in the study were randomized to Ranitidine

intervention or Omeprazole (standard therapy) for the treatment of gastroesphogeal

reflux disease. They were administered a GSRS once at baseline and once after 6

Page 31: To my parents for their unyielding support and encouragement

31

weeks of intervention (42). Mean GSRS syndrome scores of treatment responders and

treatment non-responders were compared with a t-test. All syndrome scores were

statistically different between treatment responders and non-responders after 6 weeks

of Ranitidine intervention. The largest change occurred in the reflux syndrome (net

change of -1.23 compared to baseline) as expected. The GSRS was sensitive enough

to detect a net change in syndrome scores as low as -0.25 indicating it is a useful tool in

the evaluation of gastrointestinal symptoms of GERD patients. To further evaluate the

discriminant validity of the questionnaire, Revicki et al. compared physician-assessed

symptom ratings to self-reported ratings from the GSRS. It was shown that the

questionnaire was able to rank symptom severity in a predictable manner when

compared to that of the physician assessed ratings. When GERD patients reported 2 or

3 symptoms to their physicians the GSRS syndrome scores scaled accordingly. This

data demonstrates the ability of the GSRS to detect changes in severity of symptoms

akin to physician-based interviews.

Responsiveness

Responsiveness of participants to the GSRS was measured by evaluating the

net change of treatment responders’ and non-responders’ mean GSRS scores (42).

Responders to Ranitidine therapy experienced a net decrease in mean reflux syndrome

scores of 1.23 compared to a decrease of 0.46 using standard therapy (p<0.0001). This

indicates that the GSRS is sensitive enough to detect changes in reflux syndrome

scores in response to differing GERD treatments. The conclusion Revicki et al. drew

was that the responsiveness measured can be used in populations presenting with

other gastrointestinal disorders as well as those with GERD, though they acknowledge

Page 32: To my parents for their unyielding support and encouragement

32

that more validation studies should be conducted to ensure its responsiveness in those

populations (42).

The GSRS is an easy to understand self-administered questionnaire that

evaluates a variety of gastrointestinal symptoms. This questionnaire has to date been

validated and used in a number of gastrointestinal disorders including irritable bowel

syndrome, gastric ulcers, and GERD. However, its use has not extended beyond sick

populations. Traditionally, symptom questionnaires are useful for detecting a reduction

in symptom severity or duration and so their use in clinical trials is encouraged, but

gastrointestinal symptoms are not exclusively experienced in sick or diseased

populations. Healthy individuals can also experience gastrointestinal symptoms typically

due to changes in diet and so, if the GSRS is able to detect decreases in syndrome

scores, it should, in practice, be able to detect increases in syndrome scores in healthy

populations. However, to date, no studies have evaluated the validity of the GSRS in

healthy populations.

Determination of Healthy Status in Research Participants

The composition of a study’s participants has an impact on the interpretation of a

study’s findings. For example, a study that evaluates the efficacy of a new cancer

treatment would be meaningless if the new treatment was not tested in cancer patients.

This example demonstrates the necessity of having a well-defined population during a

clinical trial. When study populations are chosen well, the study’s conclusions have a

higher degree of validity because results are not being affected by external variables.

External variables that can alter data are called confounders or confounding variables

(38). Scientists control for confounding variables through study design. In clinical trials,

study participants are usually the largest source of confounding variables simply due to

Page 33: To my parents for their unyielding support and encouragement

33

the fact no human is physically or socially identical to another. A method by which study

participants can be controlled for is by defining inclusion and exclusion criteria for study

volunteers. Inclusion and exclusion criteria in phase I clinical trials evaluating probiotics

aim to prevent the active enrollment of individuals with preexisting gastrointestinal

disorders. Investigators are able to ensure that any gastrointestinal symptoms

experienced during the study are not a result of probiotic consumption by excluding

volunteers with gastrointestinal disorders from taking part in the study. Another method

used to prevent the introduction of confounders is through the use of questionnaires.

Questionnaires serve as both screening and diagnostic tools depending on the context

of their use. One such questionnaire is the Global Physical Activity Questionnaire

(GPAQ).

In a systematic review of 89 studies involving physical activity questionnaires,

Helmerhost et al. tested the reliability and validity of the GPAQ using intraclass

correlation coefficients (ICC), Pearson and Spearman correlation coefficients and

Cohen’s weighted kappa. Pearson and Spearman correlation coefficients and Cohen’s

weighted kappa are described in the GSRS section (45). The intraclass correlation

coefficients, much like Cohen’s weighted kappa, were used to measure the reliability of

a quantitative measure made by two different interviewers when a questionnaire was

not self-reported.

The Global Physical Activity Questionnaire was developed by the World Health

Organization in 2002 to identify inadequate physical activity levels, a key risk factor for

disease. The questionnaire classifies an individual’s activity level based on a

combination of its intensity (classified as vigorous, moderate, and light) and duration

Page 34: To my parents for their unyielding support and encouragement

34

(measured in minutes/week) in a typical week. Bull et al. are the main contributors to the

validation of the GPAQ and they conducted a study where over 3600 participants were

either administered or received a self-report version of the GPAQ on their first

consultation, at this time they were given a pedometer or accelerometer in order to

provide an unbiased assessment of their physical activity level. On their second and

third clinical visits, they were given the questionnaires again in order to assess reliability

and validity. Construct validity was measured by comparison with an instrument

developed by the investigators based off the International Physical Activity

Questionnaire, which has previously been validated (46). The International Physical

Activity Questionnaire is administered in a short and long form, both of which have their

own strengths and weaknesses. The short version lacks domain specific estimates and

the long version contains 27 items which is too cumbersome for administration in large

clinical trials. The GPAQ is an attempt at capitalizing on the strengths of the short (easy

to use and administer) and long (accurate estimates of duration and intensity) versions

of the international physical activity questionnaire.

The GPAQ measures physical activity levels under three domains: leisure and

recreation, work, and transportation. Activity level is defined as either moderate or

vigorous and is qualified by frequency in the past week. Bull et al. reported that

reliability coefficients presented with moderate to substantial strength, construct validity

between GPAQ and the IPAQ instruments also showed moderate to strong positive

correlations but criterion validity was only in the poor to fair range. These results are

overall very positive as the GPAQ has demonstrated consistent activity level scoring

across several different cultural regions such as Ethiopia, Indonesia, India and Japan.

Page 35: To my parents for their unyielding support and encouragement

35

Criterion validity, which objectively measures physical activity with pedometers or

accelerometers provided to participants, resulted in poor to fair agreement with self-

reported values. The investigators explained this by stating that the GPAQ should be

evaluated in more countries to decrease the influence of a particular culture or

geographic region on the data collected. The issue that Bull et al. faced with using

pedometers and accelerometers is that they only measure movement of the body and

may not accurately represent how much physical exertion a person experiences. This

diminishes the value of comparing pedometer measurements against self-reported

activity levels. Overall, the use of the GPAQ as a consistent and reliable measure of

self-reported activity level does not have the best consistency and validity. Study

participants can unknowingly over or under estimate exercise levels due to the

subjective nature of physical exertion. A study participant who only recently began

cardiovascular exercises may consider walking at a brisk pace vigorous while a

seasoned marathon runner would only consider it as light in intensity. This subjective

variability in self-reported physical activity levels can introduce recall bias into data

collected. With this in mind, the GPAQ is likely not the best tool for evaluating the

efficacy of physical activity on health conditions because more reliable and unbiased

tools, such as daily diaries, can be used for this purpose. These limitations lead to the

conclusion that the GPAQ is better suited for evaluating the perceived activity level of a

given sample and not as a diagnostic measure. This makes it an ideal screening tool.

Bacillus subtilis as a Probiotic

Bacillus subtilis is a gram positive, spore forming, bacteria that is most commonly

found in the soil (47). Bacillus spores are unique because of their resistance to stomach

acid and pancreatic juices, which increases their chances of survival through the human

Page 36: To my parents for their unyielding support and encouragement

36

gastrointestinal tract. It is hypothesized that once the spores have reached the large

intestine they begin to germinate where they can exert probiotic effects (48). Bacillus

subtilis R0179 is currently marketed throughout Asia as Medilac-DS/S®, S/DS refer to

single and double strength respectively, in a 1:9 formulation with Enterococcus faecium

R0026 (DS dosage: 6x109 CFU/d = 2 caps). This probiotic formulation has been

evaluated in approximately 100 clinical investigations and evaluated by two systematic

reviews (49). The studies, reviewed by Tompkins et al. 2010, evaluated the efficacy of

the probiotic in several different disease states. A majority of these studies were

published in the Chinese, Japanese and Korean language journals making it difficult to

directly evaluate each study. Tompkins et al. 2010 also had limited access to some

studies forcing them to rely solely on English abstracts for their review.

Tompkins et al. 2010 first report on the ability of Medilac-S® to change the

microbiota composition of cirrhotic liver patients to resemble that of healthy controls.

Fifty cirrhotic liver patients and 20 healthy volunteers were randomized to Medilac-S® or

Bifico (a probiotic containing Bifidobacterium sp.-undisclosed, Lactobacillus acidophilus

and Enterococcus sp.-undisclosed in a total dosage of 3x109 cfu/day). Intestinal

microbiota analysis revealed a significant difference at baseline that was resolved after

two weeks of intervention (2 capsules at 3x109 CFU/capsule). While this data provides

some evidence towards the impact of B. subtilis R0179 on the intestinal microbiome,

Tompkins et al. 2010 caution classifying this shift in intestinal microbiota as a health

benefit. Furthermore no data was collected evaluating the long term effects of this shift

in intestinal microbiota.

Page 37: To my parents for their unyielding support and encouragement

37

Second, Tomkins et al. 2010 report on the evaluation of Medilac-DS® use, in

conjunction with traditional therapy, to eradicate Helicobacter pylori in gastric ulcer

patients (n=352). Probiotic intervention lasted for 8 weeks. The study resulted in an

83.5% H. pylori eradication rate in the probiotic group compared to 73.3% in the control

(p=0.027). Patients randomized to the probiotic intervention also experienced less

diarrhea and fewer overall symptoms compared to traditional therapy. The investigators

believe that the decrease in total side effects resulted in an increase in drug compliance

which in turn increased eradication rates. These results are promising for the continued

use of Medilac-DS as adjuvant therapy for H. pylori eradication. The reduction of

symptoms, however, is not entirely unique to Medilac-DS. Several other studies have

demonstrated that probiotic formulations composed of Lactobacilli, Bifidobacterium and

fermented milk products like Kefir, also reduce symptoms associated with traditional H.

pylori eradication (50-52). A major limitation of these studies is their use of generic

questionnaires which have not been validated in H. pylori patients. This immediately

casts some doubt on their symptom data and its ability to adequately detect decreases

in symptom intensity and presence. The investigators caution that while there was an

increased eradication rate of H. pylori while consuming Medilac-DS® this may only be

due to an increase in compliance and not a direct result of interaction between E.

faecium, B. subtilis, and H. pylori.

Third, Tompkins et al. 2010 report on four double-blind, placebo-controlled trials

that evaluated the effect of probiotic interventions, in addition to traditional therapy, on

irritable bowel syndrome. These clinical trials resulted in significant decreases in

abdominal pain, but no reduction in gas expulsion, bloating, defecation or other

Page 38: To my parents for their unyielding support and encouragement

38

gastrointestinal symptoms commonly reported in irritable bowel syndrome patients. The

most promising of these studies was a randomized, double blind, placebo-controlled

clinical trial (n=34) which utilized Medilac-DS® (4 weeks) as mono-therapy for IBS. This

study resulted in a significant reduction in frequency (p=0.038) and severity of

abdominal pain as measured by a visual analog scale (p=0.044). The main text of the

article is published in Korean and so access to details of study design is limited.

Tompkins et al. 2010 do not report on whether frequency and severity were measured

over the entire 4-week intervention period or only in the baseline and post intervention

time point. If the reduction in frequency and severity of abdominal pain occurred only in

week 4 of intervention and not in weeks 2 or 3, then much doubt is cast on the ability of

Medilac-DS® to reduce IBS symptoms.

Fourth, Tompkins et al. 2010 report on seven clinical trials (n=745) evaluating

Medilac-DS/S® use in participants with acute, chronic, and antibiotic-associated

diarrhea. The studies yielded some positive results but Tompkins et al. could find no

reason to cite the Medilac-DS/S® probiotic interventions as the cause partially due to

study design. Only one of the studies evaluating the various diarrhea types was blinded.

This study compared the effects of Medilac S® and Pei Fei Kang® (undefined probiotic)

on acute and chronic diarrhea in a randomized, single-blind, placebo-controlled trial

(acute diarrhea: n=95, chronic diarrhea: n=48). The intervention period was 5 days and

there was no difference detected between groups. It is unclear whether the primary

outcome of this study was to treat diarrhea or to compare the effect of the different

probiotics. This study could have benefitted from a placebo arm to evaluate overall

efficacy of the probiotics. While the study was single blinded, the participants would

Page 39: To my parents for their unyielding support and encouragement

39

have still been notified that they would be randomized to a probiotic, either Medilac S®

and Pei Fei Kang®, thus diminishing the benefits of blinding. Any results demonstrating

a benefit attributed to the probiotics may be confounded due to the placebo effect.

Fifth, Tompkins et al.2010 report on ten clinical trials (n=528) evaluating the

effect of Medilac-S® in ulcerative colitis patients. These studies were randomized and

used active controls (traditional therapy) but were not blinded. The studies

demonstrated a 12%-31% increase in the treatment of ulcerative colitis when traditional

therapy was combined with the consumption of Medilac-S (3x109 CFU/day).

Intervention periods ranged from 2-12 weeks. Tompkins et al. 2010 concluded that

these studies were adequate in terms of participant selection, randomization and dosing

regimens but that they were poorly designed in terms of blinding, assessment of

compliance and statistical analyses which casts doubt on their results.

Of the studies reviewed by Tomkins et al. 2010 that reported adverse events no

serious adverse events or reactions could be linked directly to the probiotic. No cases of

bacteremia or septicemia occurred. A handful of symptoms were reported among the

various studies including nausea, vomiting, dizziness and feelings of being flustered, but

these symptoms did not persist and were isolated to only a handful of individuals that

were randomized to the probiotic interventions. Despite the formidable research on this

probiotic formulation, there are no studies which evaluate the B. subtilis R0179 strain as

a single probiotic agent (49).

Tompkins et al. 2010 commented on the validity of some conclusions drawn from

the reviewed studies. They cite poor study designs (49). Tompkins et al. 2010 also

conclude that randomized, double-blind, placebo-controlled clinical trials are necessary

Page 40: To my parents for their unyielding support and encouragement

40

before any symptom reduction or increased efficacy can be attributed to probiotic

therapy with a high degree of confidence (49). More work must also be done on the

analysis of the probiotic based on dosage levels which would, in turn, necessitate closer

monitoring and better reporting of symptoms and adverse events in the populations

being studied.

The continued acceptance and usage of probiotics in different regions of the

world has an effect on the popularity and acceptance of these products for their

therapeutic properties. Both patients and their physicians will increasingly find

themselves in situations where they need to understand whether a particular type of

probiotic supplement can have a beneficial effect on a disease state. Due to the

increasing costs of health care and the monetary and symptomatic burdens some drugs

place on patients, probiotics are continuously being turned to as alternative and

supplemental health care. It is with these reasons in mind, along with the general

understanding of safety of a particular strain, that more research needs to be

conducted. Tompkins et al. 2010 have presented some evidence that B. subtilis R0179

is a safe product when consumed with E. faecium R0026, but its use in diseased

populations must be examined more closely (47). This warrants the use of randomized,

double-blind, placebo-controlled studies that utilize validated health-related quality of life

questionnaires and microbial sequencing techniques to assess the viability of the

bacteria post ingestion. When a health benefit is suspected, molecular techniques

identifying relevant biomarkers should exist to substantiate any related improvement in

quality of life as determined by questionnaires.

Page 41: To my parents for their unyielding support and encouragement

41

In Vitro Studies

The Safety of Bacillus subtilis

The usage of the spore forming Bacillus genus of bacteria as a probiotic has

grown in popularity. A study by Sorokulova et al. conducted on Bacillus subtilis BS3,

taken from the Ukrainian Collection of Microorganisms, cultured the strain to evaluate its

safety. 16S rRNA sequencing was conducted to identify the bacteria via an identification

key. Cultured bacteria were also evaluated for antibiotic resistance, the presence of

plasmids, enterotoxin genes and virulence factor analysis including enterotoxin

detection. Adhesion toxicity studies were carried out concurrently in a murine model

(n=220) (53).

Strain BS3 was identified as a gram positive, rod shaped bacteria capable of

forming endospores with the ability to synthesize catalase, an enzyme that catalyzes

the degradation of hydrogen peroxide to water (53). Hydrogen peroxide acts as an anti-

microbial in the human gastrointestinal tract and is classified as a reactive oxygen

species. Bacteria that can neutralize reactive oxygen species have a greater chance of

surviving mammalian gastrointestinal tracts, though this trait is not desirable in

pathogenic bacteria (54). Strain BS3 lacked a plasmid and was susceptible to all

antibiotics mandated by EFSA (53) . BS3 was resistant to Oxacillin and was moderately

resistant to Amoxicillin, Methicillin and some Cephalosporins. This was not a concern to

Sorokulova et al. as Bacillus subtilis is non-pathogenic and the resistance was below

the maximally-recommended resistance concentrations as determined by regulatory

bodies. BS3 presented low adhesion rates to mucin and Caco-2 cells, no enterotoxin

genes were found after DNA analysis of the bacteria and no virulence factors (such as

the Hbl gene which is often associated with diarrhea induction) were found (53). The in

Page 42: To my parents for their unyielding support and encouragement

42

vivo acute toxicity study subjected mice (n=220) to high dosages of BS3 orally (5x107 to

2x1011 CFU/mouse) and intravenously (5x107 to 5x109 CFU/mouse). No treatment-

related deaths or declines in general health (decreased activity and weight gain)

occurred. No mice exhibited chronic toxicity symptoms (53). Although the results of this

study cannot be translated to all strains of B. subtilis, they are good examples of how

the safety of a particular bacterial strain should be evaluated before clinical trials.

Sorokulova et al. also evaluated the chronic toxicity of other B. subtilis strains in

mice, guinea pigs and rabbits (53). Ten New Zealand White rabbits were exposed to

daily oral dosages (1x109 CFU/day) of the spore form of B. subtilis for 30 days. Animals

were then sacrificed and blood was drawn and tissue samples were taken of the

visceral organs for histology. The rabbits showed no adverse symptoms or side effects

from consumption of the bacteria. Histology of the organs and lymph nodes showed no

change in bacterial content indicating no bacteremia (53). The chronic toxicity study was

also done on guinea pigs where a single dosage of spores was given at 1x109 CFU/day.

After 17 days of monitoring the, generally sensitive guinea pigs, showed no

abnormalities in appetite, behavior, feces and weight (53). No toxicity levels were

observed in either control or treatment groups.

The Safety of Bacillus subtilis R0179

The use of B. subtilis R0179 in the probiotic formulation Medilac DS/S as

described previously carries a considerable amount of evidence towards its safety for

human consumption. In an effort to further evaluate the safety of this particular strain,

Tompkins et al. 2008 conducted genetic identification, plasmid analysis, enterotoxin

analysis, antibiotic resistance, human intestinal epithelial adhesion, and acute toxicity

studies (47).

Page 43: To my parents for their unyielding support and encouragement

43

The genetic identification of strain R0179 confirmed its inclusion in the Bacillus

subtilis genus and species (47). This test was conducted with 16srRNA sequencing with

a 100% match rate to a standardized B. subtilis genome sequence. Strain R0179 lacked

plasmids and DNA analysis revealed the absence of the toxin genes hbl, nhe and cytK

when compared to Bacillus cereus (a positive control). These enterotoxins genes are

linked to the production of enterotoxins and have been implicated as the cause of B.

cereus’ classification as a food borne pathogen (55). Antibiotic resistance analysis

revealed that strain R0179 was below maximally-recommended resistance levels for all

antibiotics tested (47). Adhesion tests revealed that strain R0179 minimally adheres to

human intestinal epithelium cell models. Its adhesion rates are about 5 times lower than

those of Lactobacillus helveticus, a bacteria commonly used in fermented milk products

(56).The acute toxicity study, a 28 day study in which 5 female pathogen free rats were

fed R0179 spores (2x109 CFU/kg/day), showed no signs of toxicity, oral intolerance,

loss in body mass, decreased food consumption, or increased mortality (47).

Additionally organ mass to body mass ratio were not significantly different from control

and strain R0179 did not migrate into the rats’ livers or spleens indicating no bacteremia

or septicemia. Expectedly strain R0179 was found at high levels in the intestinal

contents of the rats (1.5x106-1.2x107 CFU/g).

Animal Studies

Germination of Bacillus subtilis in the Gastrointestinal Tract

Bacillus subtilis’ spore form confers upon it many beneficial factors like high

resistance to wet and dry heat, ultra-violet and gamma radiation, desiccation and

oxidation (57). B. subtilis is most often consumed while it is in the spore form which

provides it resistance to the acidic contents of the stomach (58). The bacteria, as a

Page 44: To my parents for their unyielding support and encouragement

44

whole, are largely aerobic though they can be induced into an anaerobic state. This

limits the viability of the bacteria in the anaerobic environment of the intestinal tract, and

so, before any probiotic effect can be attributed to B. subtilis, the mechanism of its

passage and survival in the GIT must be understood (48).

In an effort to discern the fate of ingested Bacillus spores Spinosa et al. isolated

B. subtilis MO1099 and inoculated 9 BALB/c mice at a dose of 109 spores. The mice

were then starved for 16 hours and sacrificed at different times post inoculation (58).

Two mice were inoculated in the gastrointestinal tract with the spore form of the

bacteria. The spores were found in all portions of the intestines, but diminished over a

period of three days whereupon they reached levels below the detectable limit. Spinosa

et al. concluded that the bacteria did not germinate into vegetative cells in the ileum,

jejunum or in the feces. The investigators concluded that any probiotic effect from B.

subtilis may in fact be due to the spore form of the bacteria (58). Their acute toxicity

study results are in line with the studies conducted on other B.subtilis strains by

Sorokulova et al. (53) and Tompkins et al. (47).

A study conducted by Casula et al. evaluated how spores of Bacillus, inoculated

(2x108 CFU/ml given in a 0.2 ml suspension) in one-week-old BALB/c mice (n=6),

germinated in the gastrointestinal tract (48). The bacteria were genetically modified to

strongly express an engineered ftsH-lacZ gene while in the vegetative state. The ftsH

gene codes for an ATPase (59) while the lacZ gene, commonly referred to as a reporter

gene, is derived from Escherichia coli genetic material and is useful for detection assays

(56). When the mice were sacrificed, an RNA primer for this genetically-engineered

gene was used to run rt-PCR allowing the investigators to detect spore germination in

Page 45: To my parents for their unyielding support and encouragement

45

the mouse gastrointestinal tract. This novel method, originally developed by Lysenko et

al. 1997, was supplemented by the collection of mice feces in order to evaluate the

transit viability of the inoculated Bacillus spores. The investigators were able to detect

as low as 102 vegetative cells, in the mouse intestinal tract. The vegetative bacteria

were found primarily in the jejunum of the mice and at levels that were only lower than

the inoculation concentration by a factor of ten. Investigators concluded that due to the

acidic composition of duodenal juices, germination may be inhibited in that portion of the

small intestine (48). While there may have been some germination in the duodenum,

the sensitivity of the assay used to detect the engineered gene may be to blame for the

inability to detect the vegetative cells in that portion of the intestines.

Casula et al. also observed that the highest concentration of Bacillus spores

were found 18 to 24 hours post inoculation indicating that both germination and spore

formation is occurring concurrently (48). This information, paired along with the fact that

vegetative cells were found post inoculation, led them to conclude that while Bacillus

may begin to colonize in the gastrointestinal tract some environmental factor prevents

its permanent colonization and thus it must resporulate leading to its subsequent

excretion in the feces. This may be due to its low adhesion rates as demonstrated, in

vitro, by Tompkins et al. 2008 on a model of human intestinal epithelial cells and

Sorokulova et al. in mucin and Caco-2 cell lines (47, 53). Any probiotic effect exerted

would likely occur during this brief colonization period wherein the bacteria could act

either as a competitive exclusion agent by briefly adhering to the intestinal epithelium

and thus prevent the adhesion of pathogenic bacteria or by some other probiotic

mechanism (48).

Page 46: To my parents for their unyielding support and encouragement

46

While the studies by Casula et al. and Spinosa et al. are contradictory with

regards to the ability of Bacillus spores to germinate in the gastrointestinal tract of mice,

it is important to consider the advantages of the rt-PCR assay used by Casula et al.

(48). This technique was able to detect levels of the vegetative cells far below those

typically observed with the common plating technique used by Spinosa et al. (58).

Furthermore, both studies contribute evidence towards the conclusion that B. subtilis

temporarily inhabits the gastrointestinal tract of mammals.

B. subtilis in the Human Intestinal Tract

Bacillus species have traditionally been considered inhabitants of soil due to their

spore forming capabilities. Research has shown, however, that once Bacillus spores

enter the gastrointestinal tract of mice, they can enter the vegetative state and

resporulate (48). However, this had not been previously demonstrated in humans (48).

Hong et al. hypothesize that Bacillus species may be permanent residents of the human

gastrointestinal tract and are only more commonly found in soil due to their spore

forming capabilities (60). The theory is that once Bacillus spores germinate in the small

intestine of man and animal they begin to outgrow their environment. As the vegetative

cells progress into the colon they resporulate where they are excreted along with feces.

This leads to their accumulation in soils. This theory is supported by evidence of

Bacillus spores being recovered from human feces (61) as well as their historical

identification as soil organisms (62).

To further elucidate the nature of Bacillus subtilis residence in the human

gastrointestinal tract Hong et al. conducted a study in healthy human volunteers. They

first retrieved fecal samples from study participants (n=29) who had not previously taken

any probiotic within the past year. Next, they retrieved two biopsies from the proximal

Page 47: To my parents for their unyielding support and encouragement

47

ileum of six ileostomy patients (60). The ileostomy patients did not have a history of

antibiotic, probiotic or laxative use at least one month before surgery. Hong et al.

reported that Bacillus spores were identified in the fecal samples collected from both

sets of participants (60). They estimated that human feces may carry on average 104

spores per gram of feces while the small intestine may carry 109 spores per gram or

greater. Hong et al. acknowledge that they did not control for extraneous sources of

Bacillus species found in food and so this could contribute to inflated estimates.

After isolating B. subtilis from the fecal and ileostomy samples, Hong et al.

proceeded to identify some key characteristics of the bacteria which may contribute to

their ability to live as gut commensals. The isolates were confirmed to grow and

sporulate under anaerobic conditions which is useful for the bacteria as they move into

the large intestine (60). It was also observed that the B. subtilis isolates were able to

form biofilms which serve to protect vegetative cells from environmental stressors like

antimicrobials and gastric juices. The biofilm has also been shown to stimulate

repopulation of the vegetative cell (63). Hong et al. conclude that a diverse community

of Bacillus species reside in the human gastrointestinal tract and that B. subtilis is likely

a gut commensal, but their total impact on the intestinal microbiome remains a mystery

(60).

Potential Health Benefits of Bacillus subtilis

The Effect of B. subtilis on Immune Function

A popular strain of the Bacillus subtilis is used in Japan for the fermentation of

cooked soybeans (called Natto) and has been shown to have probiotic properties (58).

This process of fermentation has resulted in the unique naming of Bacillus subtilis as B.

natto or B. subtilis var. natto (58). The effect of B. natto on the B and T lymphocytes in

Page 48: To my parents for their unyielding support and encouragement

48

the spleens of chickens was investigated in 1986 by Inooka et al. Chicks were fed B.

natto supplemented feed (107 cfu/day) for 27 days and then sacrificed. Analysis of T

and B lymphocytes in the spleens showed that chicks fed the supplemented feed had

more T and B lymphocytes compared to those on a normal diet. This suggests that the

B. natto strain has some effect on immune functions in chicks (64).

Popular probiotics which have well established probiotic effects, such as

immunostimulatory properties, are Lactobacillus species (65). An in vitro study,

conducted by Hosoi et al. investigated the effect of B. subtilis natto on the viability of

human derived Lactobacillus reuteri and Lactobacillus acidophilus strains that were

independently co-cultured with B. subtilis in an aerobic environment (66). Hosoi et al.

observed an increase of viable cells of both L. reuteuri (4 log increase) but not in L.

acidophilus (1 log decrease) when co-cultured with B. subtilis for 6 days when

compared to independently cultured Lactobacilli spp. This positive relationship extended

beyond enhanced growth. When toxic levels of hydrogen peroxide were added to the

cultures, B. subtilis natto was able to prolong the life of L. reuteri by three days whereas

L. acidophilus cells perished much faster than independently co-cultured samples. This

increased viability was hypothesized to be a result of the enzyme subtilisin, produced by

B. subtilis which catalyze the degradation of hydrogen peroxide to water similar to many

other catalases. The study shows promise due to its intention to link growth promoting

properties B. subtilis (natto) with that of other probiotics. Of important note is the fact

that this particular B. subtilis (natto) strain is used in the manufacture of food products in

Japan (66). Alongside this is the fact that the investigators used human derived

Lactobacilli strains which indicates that the effect may also be observed in vivo (66).

Page 49: To my parents for their unyielding support and encouragement

49

Although this study provides evidence towards the conclusion that B. subtilis has a

positive impact on the viability of certain Lactobacilli strains it does not acknowledge

that these benefits may not impact the human intestinal microbiome on a biologically

significant scale.

The Effect of B. subtilis on Bone Metabolism

In previous studies, Tsukamoto et al. established that B. subtilis (natto) had a

positive impact on the production of Vitamin K2, a vitamin has been shown to take part

in the prevention of bone loss particularly in older populations suffering from a decline in

bone density (67). Vitamin K2 plays a role in the carboxylation of osteocalcin which is an

osteoblast protein used in the formation of bone matrix and is found abundantly in natto

a food commonly eaten in Japan. When the natto has been fermented with B. subtilis

vitamin K2 levels increase from 775µg/100g of natto to 1298µg/100g of natto (67). To

evaluate the effect of natto Tsukamoto et al. conducted a study (n=134) to evaluate the

effects of natto consumption on serum vitamin K2 and osteocalcin levels. This study was

not a direct consumption of B. subtilis, but rather an example of how B. subtilis can be

utilized as a functional component in food production.

Results of the study demonstrated that people who fell into a category of

occasionally (a few times a month) or frequently (a few times a week) consuming natto,

supplemented with B. subtilis, had significantly higher levels of serum vitamin K2

compared to those that only consumed natto rarely (less than a few times a month)

(67). This is coupled with evidence of higher serum levels of osteocalcin present in the

occasional and frequent groups indicating higher bone repairing activity. People who

frequently ate natto had vitamin K2 levels increase by us much as 2.5 ng/ml, while

osteocalcin only increased in men by as much as approximately 3 ng/ml. This study

Page 50: To my parents for their unyielding support and encouragement

50

demonstrates great promise as a method by which B. subtilis can be utilized as a

probiotic agent.

The Use of B. subtilis as Bowel Preparation for Colonoscopy

Colonoscopy viewing is a medical procedure used to evaluate the presence of

colonic lesions in humans which can develop into colon cancer. To prepare for the

procedure, patients are asked to consume either polyethylene glycol or sodium

phosphate solutions (68). The solutions act to cleanse the colon but patient non-

compliance and other indicators, such as constipation, obesity and cirrhosis of the liver,

can result in unsuccessful viewings of the colon (68). This can result in cancelled

procedures, and added medical cost. In an effort to determine whether the consumption

of Medilac ®, as co-treatment to polyethylene glycol or sodium phosphate solutions,

improved the efficacy of the traditional colon cleansing therapies, Lee et al. conducted a

randomized, single-blinded, active control, clinical trial (n=107).

Patients who presented with constipation as defined by Rome III criteria were

enrolled in the study in parallel to controls that were not constipated but still scheduled

for colonoscopy. For two weeks, the participants who received the probiotic intervention

consumed one capsule orally three times per day. On the night before and on the

morning of the colonoscopy, they consumed the traditional sodium phosphate solution.

Colonoscopy preparation was evaluated immediately after each procedure by

endoscope. The group of constipated patients that received the probiotic had fewer

unsatisfactory scores compared to the constipated patients that received placebo

(45.1% vs 79.2% p<0.0001). There was no difference when comparing normal patients

with constipated ones. Interestingly, the group that received probiotics had lower

incidences of vomiting and bloating related adverse events.

Page 51: To my parents for their unyielding support and encouragement

51

The study merits attention as its use of probiotics was paired to enhance a

medical procedure’s effectiveness. The study, however, failed to evaluate symptoms

due to probiotic consumption there may have been a myriad of symptoms that

participants experienced while taking the probiotic that were not recorded. The group

that received the probiotic in the constipated group showed a significantly better total

colon cleansing score when compared to placebo, with the assumption that this is due

to alleviation of the constipation by the probiotic, however no effort was made to record

bowel movement frequency in participants (69). If Lee et al. had used a questionnaire to

evaluate stool frequency, consistency and gastrointestinal symptoms, the impact of B.

subtilis could have been further elucidated. At this point in time, no research evaluating

the effect of Bacillus probiotics in constipated patients has been conducted.

Conclusions

The increasing acceptance and usage of probiotics in different regions of the

world is due to the current state of research which seeks to better understand the

impact of probiotics on human health. Both patients and their physicians are

increasingly finding themselves in situations where they need to decide if a particular

type of probiotic supplement can have a beneficial impact on a disease state. Due to the

increasing costs of health care, and symptoms associated with certain drugs, probiotics

are continuously being turned to as alternative and supplemental health care. It is with

these reasons in mind that the study of probiotics requires a multidisciplinary approach

that evaluates both the microbiological and clinical impacts of probiotic consumption.

This warrants the use of randomized, double-blind, placebo-controlled studies that

utilize validated, health-related quality of life questionnaires, microbial sequencing

techniques and metabolomics to substantiate causal relationships between probiotic

Page 52: To my parents for their unyielding support and encouragement

52

usage and health benefits. This type of research will increase the confidence in which

probiotics can be turned to as adjuvant and alternative therapies for disease states.

Table 1-1. Syndrome assignments of the SF-36, PGWB, and GSRS

SF-36 PGWB GSRS

Physical fFunction

Role Limitations-Physical

Bodily Pain

General Health

Mental Health

Role Limitations-Emotional

Vitality

Social Function

Mental Component Summary

Physical Component Summary

Anxiety

Depression

Positive Well-Being

Behavioral Control

General Health

Vitality

Abdominal Pain

Reflux

Indigestion

Constipation

Diarrhea

Page 53: To my parents for their unyielding support and encouragement

53

CHAPTER 2 PURPOSE

The purpose of this study is to evaluate the effect of the probiotic B. subtilis

R0179 on gastrointestinal function, general wellness and gastrointestinal viability in

healthy young adults. To date, no studies have been conducted evaluating the dose

response of a probiotic on gastrointestinal function, general wellness and

gastrointestinal viability. The current study aims to fill this gap in the literature by

administering oral doses of the probiotic B. subtilis R0179 to healthy adults for a period

of 4 weeks and to evaluate the development of any general wellness or gastrointestinal

symptoms through the use of a daily questionnaire and the validated gastrointestinal

symptom response scale. It is hypothesized that B. subtilis R0179 will have no effect on

the symptoms reviewed in the questionnaires and that due to its spore forming

capability, B. subtilis R0179 will survive the antimicrobial properties of the human

gastrointestinal tract and pass through to the colon where it can exert probiotic effects.

Page 54: To my parents for their unyielding support and encouragement

54

CHAPTER 3 METHODS AND PROCEDURES

Study Design

A six-week, randomized, double-blind, placebo-controlled study was carried out

with 81 healthy adults, 18-50 years of age. Participants were started on the protocol in

five waves consisting of a one week baseline period, a four week intervention period

and a one week washout period (Figure 3-1). Wave 1 was randomized July 30th 2012

(n=21), wave 2 was randomized August 6th (n=17), wave 3 was randomized August 13th

(n=20), wave 4 was randomized August 20th (n=19), and wave 5 was randomized

August 27th (n=4). All participants were randomized in parallel into one of three possible

dosages of the probiotic B. subtilis R0179 (10 billion, 1 billion and 0.1 billion

CFU/capsule) or to the placebo group. Institutional Review Board approval was

obtained from the University of Florida’s IRB-01 on 06/28/2012 for a period of one year

ending on 06/19/2013, this was extended to 6/20/2014 for further data analysis

(Appendix A).

Inclusion and Exclusion Criteria before Attaining Consent

Participants were included in the study if they fell within the age range of 18-50

years. They were required to be willing and able to complete the Informed Consent

(Appendix B) in English as well as completing a Global Physical Activity Questionnaire

(GPAQ) and being determined moderately active by the GPAQ (Appendix C).

Moderately active was defined as having to complete at least sixty minutes and at most

three hundred minutes of moderate to vigorous intensity exercise(s) per week. Any

participants who fell outside of these parameters were excluded from participating in the

study. These parameters were selected as recommended by the American College of

Page 55: To my parents for their unyielding support and encouragement

55

Sports Medicine in a position stand published in 1998 (70) Participants were required to

be willing to take height and weight measurements as well as providing demographic

information. They were required to be willing to consume the B. subtilis R0179 (at

approximate doses of 0.1, 1.0, and 10 billion CFU/capsule/day) or a placebo for a 28

day period. They were required to have Internet access for the duration of the study in

order to complete a daily questionnaire electronically (Appendix D). They were also

required to complete the Gastrointestinal Symptom Response Survey (GSRS) three

times throughout the study: once in the baseline week, once in week 4 of the

intervention period and once after the washout period (Appendix E). In addition, they

were required to provide three stool samples (baseline, week 4 of intervention, and

during washout).

Participants were excluded from the study if they did not meet any of the above

criteria and if they were taking any medications for constipation or diarrhea. They were

excluded if they had taken antibiotics within the past four weeks prior to randomization

or if they were already taking a probiotic supplement that they did not want to

discontinue at least two weeks prior to the beginning of the study. In addition, they were

excluded if they had or were currently being treated for any diseases or illnesses such

as the gastrointestinal diseases: gastric ulcers, Crohn’s disease and ulcerative colitis or

other chronic diseases such as diabetes and kidney disease or immune-compromising

diseases/conditions such as HIV, AIDS, hepatitis, and cancer, or if they were a

transplant patient.

Recruitment

Healthy adults were recruited by flyers and word of mouth on the University of

Florida campus. Following an initial screening of participants using the inclusion and

Page 56: To my parents for their unyielding support and encouragement

56

exclusion criteria listed above, participants were consented by trained study

coordinators and proceeded to take the GPAQ. After completion of the GPAQ, any

participant that completed less than sixty and more than three hundred minutes of

vigorous to moderate intensity exercise per week were notified of their ineligibility.

Recruitment began on 7/02/2012 and ran intermittently until 08/23/2012.

Baseline

Following informed consent, participants were provided with a customized web

URL to their daily questionnaires which were to be completed in the evening before

11:59pm of that day. Participants were also instructed on the proper technique for the

collection of a whole stool sample by trained research staff. They were provided a

commode specimen collection system (Fisher Scientific) and were instructed to bring

the sample in for processing on ice within four hours of defecation. Following the

completion of the baseline week, participants attended their first clinic visit in which

research coordinators administered the GSRS, took weight measurements on a Health

o meter® Professional scale (model: 349KLX) and height measurements on a Seca

Stadiometer (model 213). Participants also completed a demographic information

questionnaire evaluating race and ethnicity. Participants were then randomized to one

of three B. subtilis probiotic groups (0.1, 1 and 10 billion CFU/capsule/day) or placebo.

Trained research staff monitored the completion of the daily questionnaires for

each participant. If the questionnaire had not been completed by 11:59 p.m., an email

reminder was sent to the participant to ensure full compliance to study protocol.

Participants who failed to comply with study protocol were withdrawn from the study

prior to randomization.

Page 57: To my parents for their unyielding support and encouragement

57

Randomization and Intervention

A simple randomization of four intervention groups was completed using

Microsoft Excel 2010 (Version 14.0.6123.5001) by a collaborator who did not have

access to study participants or data. No restrictions were implemented in the

randomization sequence. Each random allocation was formatted to fit on a single sheet

of white paper, indicating the coded intervention group, the date, the number of pill

bottles given to the participants and the participant identification number. These were

placed in envelopes and sealed to ensure the integrity of the blinding process. The

envelopes were separated into five stacks, each stack represented a different

randomization date, on top of each envelope was printed: Stack # and Envelope #.

Participants were enrolled by trained research coordinators during the consenting

process. On randomization day research coordinators unsealed the envelopes and

assigned the participants to their intervention group.

Post randomization, participants were instructed to take one capsule per day at

the end of a meal. They were provided with enough capsules distributed between two

pill bottles for the entire 4-week intervention period. The bottles were clearly marked

with a specialized four digit code (for the purposes of randomization) and the

instructions, “Consume one capsule per day at the end of a meal.” Participants were

then instructed on how to open the bottles and that they should begin consumption of

the capsule that day (Day 8 of the study) and to continue doing so for four weeks. They

were also instructed to retain the bottles and any extra capsules. In the last week of the

intervention phase, participants provided their second fecal sample.

All research staff were blinded. Capsules provided by Lallemand Health

Solutions (Montreal, QC) were identical in size, shape and color. The capsules from

Page 58: To my parents for their unyielding support and encouragement

58

each intervention group were inside white, unmarked, and sealed pill bottles. Labels for

each intervention group were applied by a separate research lab in order to ensure the

fidelity of the blinding process.

Washout and Post Intervention

Following the intervention phase, participants attended their second clinical visit

where they returned the pill bottles and any remaining capsules. They were

administered a GPAQ and GSRS by study coordinators. Final weight measurements

were taken. Participants were then instructed to continue completing their daily

questionnaires for the next seven days and that they would be administered a final

GSRS on the last day of the washout period. They were also instructed to bring in a

final stool sample immediately following the washout week.

Compensation

Following completion of the entire study: six weeks of completing the daily

questionnaire, consumption of the capsules, and providing three fecal samples

($15USD/stool sample) participants were provided a maximum of $200USD.

Stool Protocol

Stool samples were collected by a commode specimen collection system (Fisher

Scientific) during baseline, intervention (week 4) and post washout to assess the

viability of B. subtilis R0179. Participants were instructed to place samples upon ice

immediately after defecation and to deliver samples to study personnel within four

hours. Samples were then homogenized by kneading in a strong plastic bag.

Approximately 1 g of homogenized stool was added to pre weighed 15 mL conical

tubes. Exact stool weight was obtained by difference and a 1:10 w/v dilution with

phosphate buffered saline (PBS) was agitated with 4 to 5 glass beads (1mm) to break

Page 59: To my parents for their unyielding support and encouragement

59

up particles. Samples were centrifuged (Sorval legend RT+) for 2 minutes at 54 rcf to

pellet large particles. Supernatants were gently mixed and 10-fold serial dilutions in PBS

were placed in a water bath at 80°C for 10 minutes to eliminate non spore-forming

organisms. Positive controls demonstrated no loss of viability due to heating. Dilutions

were plated using in duplicate on Luria Bertoni agar (Fisher Scientific, #BP1425-500)

and incubated overnight at 37°C. Spreaders were sterilized by incineration to prevent

contamination from spores.

Statistical Methods

Equivalence Testing

Equivalence testing was conducted for both the Daily Questionnaires (DQ) and

the GSRS. Syndromes and symptoms of the daily questionnaires were tested for

unequal variances using Levene’s test, upon rejection of the null hypothesis (α =0.05)

data was transformed accordingly. Each syndrome of the daily questionnaire was

compared on a between groups and per week basis, with a predetermined specified

practical difference threshold of .377 (approximating a value of one unit on the

questionnaire) after the data was transformation. When mean symptom and syndrome

scores were significant (P<0.05), the null hypothesis was rejected indicating a practical

equivalence between the intervention groups. Each syndrome of the GSRS was

compared on a between group and per intervention period basis, with a predetermined

specified practical difference threshold of 1. When mean syndrome scores were

significant (P<0.05), the null hypothesis was rejected indicating a practical equivalence

between the intervention groups.

Daily Questionnaires evaluated a variety of general wellness symptoms (Table 3-

1 and Table 3-2) that were rated on a scale of 0 (no symptoms present) to 6 (very

Page 60: To my parents for their unyielding support and encouragement

60

severe). Means of subject responses were taken for each symptom by week. These

weekly means were then grouped with other similar symptoms to form a mean

syndrome score. The syndromes analyzed were: GI distress, cephalic, ear-nose-throat,

behavioral, emetic, and epidermal. Symptoms that could not be grouped into a

syndrome were evaluated individually. Due to non-normality, square roots of syndrome

scores were taken.

Individual symptoms of the DQ that did not fit into a syndrome grouping were

also tested for equivalence using transformed data. Mean sleep equivalence testing

was conducted at a predetermined specified difference threshold of 30 mins. Mean

bowel movements was tested for equivalence at a predetermined specified practical

difference threshold of .20 bowel movements, bowel movements were pooled into

baseline, intervention (weeks 1-4) and washout scores. Kruskall-Wallis tests were

conducted to evaluate the change in syndrome and symptom scores within intervention

groups over the 6-week study. When significance was reached the pair-wise

comparisons were made using the Wilcoxon test.

Gastrointestinal Symptom Rating Scales were administered to participants at

baseline, week 4 of intervention and post washout. Individual symptoms assessed by

the GSRS were averaged into syndrome scores (Table 3-2), these syndrome scores

were then compared across the baseline, intervention and washout periods between

groups. Kruskall-Wallis tests were conducted to evaluate the change in syndrome

scores within intervention groups over the baseline, intervention and washout periods.

When significance was reached the pair-wise comparisons were made using the

Wilcoxon test.

Page 61: To my parents for their unyielding support and encouragement

61

B. subtilis Viability

Viability of B. subtilis R0179, recovered from stool samples, was assessed using one

way ANOVA followed subsequently by Tukey-Kramer HSD when significance was

reached (p<0.05). Data was normalized when appropriate. Data analyzed was log

(CFU/g).

Page 62: To my parents for their unyielding support and encouragement

62

Figure 3-1. Study design

Page 63: To my parents for their unyielding support and encouragement

63

Table 3-1. Daily Questionnaire syndrome assignments

Gastrointestinal Distress

Cephalic ENT Behavioral Emetic Epidermal Unclassified symptoms

Bloating Headache Sore throat Anxiety Nausea Itching Fatigue

Flatulence Dizziness Runny eyes Depression Vomiting Skin rash Satiety

Stomach noises Nasal congestion Skin redness and flushing

Hours of sleep

Abdominal cramps Blocked ear canal

Constipation

Diarrhea

Bowel movement frequency

Page 64: To my parents for their unyielding support and encouragement

64

Table 3-2. Gastrointestinal Symptom Rating Scale syndrome assignments

Abdominal Pain Indigestion Reflux Constipation Diarrhea

Abdominal pain Stomach rumbling Heart burn Constipation Diarrhea

Hunger pain Bloating Acid regurgitation Hard stools Loose stools

Nausea Burping

Increased Flatus

Feelings of incomplete evacuation Urgent need for defecation

Page 65: To my parents for their unyielding support and encouragement

65

CHAPTER 4 RESULTS

Participants

Eighty-one participants completed the six week study (Figure 4-1). One

participant in the 10 billion CFU intervention group voluntary withdrew after experiencing

some itching and redness symptoms and another was withdrawn from the study due to

non-compliance to study protocol. One serious adverse event was reported by a

participant randomized to the 0.1 billion CFU/capsule/day intervention, but this

participant did not cease consumption of the probiotic and completed the study.

Participant ages ranged from 20 to 49 across all intervention groups and did not differ in

gender, age, race/ethnicity, BMI categories, physical activity level, questionnaire

compliance and intervention compliance (% total of capsules consumed during

intervention). The 0.1 billion CFU/group differed from the other intervention groups in

the stool compliance category (85% compliance vs. 100, 97 and 90 in the placebo, 0.1

and 10 billion CFU groups respectively, p<0.01) (Table 4-1).

Daily Questionnaire Analysis

Daily Questionnaires (DQ) were administered to participants each night for the

duration of the study, a total of 3402 DQ were completed and analyzed. Syndrome

testing is presented in Table 4-2. Individual symptom data is presented in Table 4-3.

The scores of the gastrointestinal distress syndrome of the DQ (symptoms: bloating,

flatulence, stomach noises and abdominal cramps) of the placebo, 0.1 billion and 10

billion CFU groups were concluded equivalent. The 0.1 billion CFU (0.3±0.1) group was

concluded not equivalent to the 1 billion (0.6±0.1) and 10 billion 0.7±0.1) CFU groups in

the baseline week. A subsequent Kruskall-Wallis test revealed that intervention groups

Page 66: To my parents for their unyielding support and encouragement

66

were no different from their mean baseline syndrome scores as the study progressed.

This indicates that intervention scores did not change over time and because all scores

were below the clinically significant threshold of 1 on the questionnaire, the syndrome

scores do not indicate that study participants were negatively reacting to consumption of

B. subtilis R0179 (Figure 4-2).

Similar results were produced for the cephalic syndrome scores (symptoms:

headache, dizziness) when non-equivalence in weeks 2 and 3 of intervention period

were concluded between the 0.1 billion CFU group (0.2±0.1) and the 10 billion CFU

group (0.3±0.1) and between the 0.1 billion CFU group (0.1±0.03) and the placebo

group (0.4±0.1) respectively (Table 4-2). Once again these scores were well below the

clinically significant threshold of 1 (Figure 4-3). A Kruskall-Wallis test confirmed that

intervention group means did not differ from baseline when compared to the intervention

and washout weeks.

The epidermal syndrome scores (symptoms: itching, skin rash, skin

redness/flushing) were concluded equivalent across all intervention by period

comparisons, means ranged from 0.02±0.02 in the 10 billion CFU group to 0.1±0.1 in

the 0.1 billion CFU group (Table 4-2). All syndrome score means were well below the

clinically significant threshold of 1 (Figure 4-4). A Kruskall-Wallis test confirmed that

group means did not differ from baseline when compared to the intervention and

washout weeks.

The ear-nose-throat syndrome scores (symptoms: sore throat, runny eyes, nasal

congestion, blocked ear canal) were concluded equivalent across all period by

intervention group comparisons except for during week 1 of the intervention period

Page 67: To my parents for their unyielding support and encouragement

67

where the 0.1 billion CFU group (0.4±0.1) and the 1 billion CFU group (0.1±0.03) groups

were concluded not equivalent (Table 4-2). All syndrome score means were below the

clinically significant threshold of 1 (Figure 4-5). A Kruskall-Wallis test confirmed that

group means did not differ from baseline when compared to the intervention and

washout weeks.

The behavioral syndrome scores (symptoms: anxiety and depression) were

concluded not equivalent for a variety of intervention and period comparisons (Table 4-

2). Scores ranged from 0.1±0.1 in the 0.1 and 1 billion CFU groups to 0.6±0.2 in the

placebo group. All mean syndrome scores were well below the clinically significant

threshold of 1 (Figure 4-6). A Kruskall-Wallis test confirmed that group means did not

differ from baseline when compared to the intervention and washout weeks.

Emetic syndrome scores (symptoms: nausea and vomiting) were concluded

equivalent across all period by intervention comparisons (Table 4-2). Mean scores

ranged from 0.003±0.003 in the 0.1 billion CFU group to 0.1±0.1 in the placebo, 1 billion

and 10 billion CFU groups. These values were well below the clinically significant

threshold of 1 (Figure 4-7). A Kruskall-Wallis test revealed a difference in the 0.1 billion

CFU group, Χ2 (5, N = 21) = 13.3, p = 0.02. Subsequent analysis using the Wilcoxon

method revealed that weeks 2 (p=0.008, 0.1±0.04) and 3 (p=0.02, 0.02±0.02) of the

intervention period and the washout week (p=0.04, 0.1±0.1) were significantly different

from week 1 (0.003±0.003) and that week 4 (0.1±0.03) was significantly different from

weeks 3 (p=0.03) and 2 (p=0.01) (Table 4-2).

Individual symptoms that could not incorporated into a syndrome score were also

tested using the equivalence testing method. The constipation symptom scores were

Page 68: To my parents for their unyielding support and encouragement

68

concluded non-equivalent for a variety of period and intervention comparisons (Table 4-

3) Scores ranged from 0.1±0.04 in the placebo group to 0.7±0.3 in the 10 billion CFU

group. All mean constipation scores were below the clinically significant threshold of 1

(Figure 4-8). A Kruskall-Wallis test confirmed that group means did not differ from

baseline when compared to the intervention and washout weeks.

The fatigue symptom also had a variety of non-equivalent period by intervention

group comparisons (Table 4-3). Mean scores ranged from 0.4±0.1 in the 0.1 billion

group to 1.1±0.3 in the placebo group. The placebo group also had a score of 1.0±0.3

during week 3 of the intervention period (Figure 4-9). A Kruskall-Wallis test confirmed

that group means did not differ from baseline values when compared to the intervention

and washout weeks.

The diarrhea symptom scores were concluded equivalent for the baseline week,

and weeks 1 and 2 of the intervention period. Means were concluded not equivalent

between the 1 billion CFU group (0.4±0.1) and the other intervention groups during

week 3 of intervention. During week 4 the 0.1 billion CFU group (0.2±0.1) and the 10

billion CFU group (0.1±0.1) were concluded equivalent; they were concluded not

equivalent when compared to the placebo (0.4±0.1) and 1 billion CFU (0.3±0.1) groups.

During the washout week, the 0.1 billion CFU group (0.1±0.04) was not equivalent to

the other intervention groups mainly because its mean was lower than the others (Table

4-3). All diarrhea symptom scores were below the clinically significant score of 1 (Figure

4-10). A Kruskall-Wallis test confirmed that group means did not differ from baseline

when compared to the intervention and washout weeks.

Page 69: To my parents for their unyielding support and encouragement

69

Bowel movement frequency was recorded daily using the daily questionnaire.

Mean bowel movement frequency ranged from 1.4±0.1 in the placebo group to 1.6 in

the 10 billion CFU group (Figure 4-11). A variety of period and intervention group

comparisons were concluded not equivalent (Table 4-3). A Kruskall-Wallis test

confirmed that bowel movement frequency did not significantly change when comparing

baseline means to intervention and washout values.

Hours of sleep were compared using the equivalence testing method. Means

ranged from 6.5±0.2 in the placebo group to 7.3 in the 1 billion CFU group (Figure 4-

12). A variety of period and intervention group comparisons were concluded not

equivalent (Table 4-3). A Kruskall-Wallis test confirmed that mean hours of sleep did not

significantly change when comparing baseline means to intervention and washout

values.

Gastrointestinal Symptom Rating Scale Analysis

A total of 243 GSRS questionnaires were completed by participants. The

abdominal pain syndrome (symptoms: abdominal pain, hunger pains and nausea)

(Figure 4-13), reflux syndrome (symptoms: heart burn and acid regurgitation) (Figure 4-

14), diarrhea syndrome (diarrhea, loose stools and urgent need for defecation) (Figure

4-15), indigestion syndrome (stomach rumbling, abdominal distension, eructation and

increased flatus) (Figure 4-16), constipation syndrome (constipation, hard stools and

feeling of incomplete evacuation) (Figure 4-17) were significant across all intervention

groups and periods indicating equivalence. Equivalence testing for the GSRS

questionnaire is presented in Table 4-4.

Page 70: To my parents for their unyielding support and encouragement

70

Viability of B. subtilis R0179 in Humans

Least square means estimates of enumerate plate counts of B. subtilis showed a

dramatic increase in the amount of viable bacteria between weeks 1, (baseline), 5

(intervention) and 6 (washout) (Table 4-5). All intervention groups showed significant

increases in bacteria counts presented as log CFU/g +1 and the placebo group was not

significant (Figure 4-18). Increases occurred in week 5, the end of the intervention

period, and diminished by the final fecal collection when probiotic consumption ceased.

Page 71: To my parents for their unyielding support and encouragement

71

Figure 4-1. Participant flow diagram

Page 72: To my parents for their unyielding support and encouragement

72

Table 4-1. Participant demographics

Placebo (n=20)

0.1 billion CFU (n=21)

1 billion CFU (n=20)

10 billion CFU (n=20)

P Value1

Gender (M/F), n 9/11 8/13 12/8 10/10 NS

Age, years Median (range) 23 (20-46) 23 (20-49) 22 (20-31) 23 (19-46) NS

Race/ethnicity2,n (%) NS

Asian 2 (10) 7 (33) 2 (10) 2 (10)

Black 4 (20) 2 (10) 2 (10) 2 (10)

Hispanic 3 (15) 2 (10) 3 (15) 2 (10)

White 9 (45) 9 (43) 13 (65) 14 (70)

Other 2 (10) 1 (4) 1 (5) -

BMI percentiles, n (%) NS

<85th (healthy weight) 10 (50) 12 (60)3 13 (65) 9 (45)

85th-94th (overweight) 6 (30) 7 (35) 4 (20) 9 (45)

≥95th (obese) 4 (20) 1 (5) 3 (15) 2 (10)

Activity Level ± SEM NS

Baseline 185.5 ± 15.9 197.3 ± 18.1 168.3 ± 16.7 199.0 ± 13.0

Washout 161.8 ± 19.9 206.3 ± 36.1 178.5 ± 28.8 234.0 ± 29.8

Compliance (%)

Questionnaire Protocol 99 98 99 97 NS

Intervention Protocol 95 89 94 93 NS

Stool Protocol 100 97 85* 90 0.01

Page 73: To my parents for their unyielding support and encouragement

73

Table 4-2. Daily Questionnaire syndrome equivalence testing

Syndrome Period 0.1 billion CFU 1 billion CFU 10 billion CFU Placebo

Gastrointestinal Distress

Baseline 0.3 (± 0.1) a 0.6 (± 0.1)

b 0.7 (± 0.1)

bc 0.5 (± 0.8)

abc

Week 1 0.4 (± 0.1) a 0.6 (± 0.1)

b 0.5 (± 0.1)

abc 0.5 (± 0.1)

abc

Week 2 0.3 (± 0.1) a 0.6 (± 0.1)

b 0.6(± 0.1)

b 0.6 (± 0.1)

b

Week 3 0.3 (± 0.01) a 0.7 (± 0.1)

b 0.7 (± 0.1)

bc 0.6 (± 0.1)

bc

Week 4 0.3 (± 0.1) a 0.7 (± 0.1)

b 0.5 (± 0.1)

bc 0.6 (± 0.1)

bc

Washout 0.3 (± 0.1) a 0.6 (± 0.1)

b 0.4 (± 0.1)

abc 0.5 (± 0.1)

bc

Cephalic Baseline 0.2 (± 0.1) 0.2 (± 0.04) 0.2 (± 0.1) 0.2 (± 0.1)

Week 1 0.2 (± 0.1) 0.1 (± 0.04) 0.2 (±0.1) 0.2 (± 0.1)

Week 2 0.2 (± 0.1) a 0.1 (± 0.04)

ab 0.3 (±0.1)

c 0.2 (± 0.1)

abc

Week 3 0.1 (± 0.03) a 0.2 (± 0.1)

ab 0.3 (±0.1)

abc 0.4 (± 0.1)

bc

Week 4 0.2 (± 0.1) 0.2 (± 0.1) 0.2 (±0.1) 0.2 (± 0.1)

Washout 0.3 (± 0.1) 0.2 (± 0.1) 0.2 (±0.1) 0.2 (± 0.1)

Data presented as mean (± std. error). Intervention by week combinations not connected by the same letter were concluded as not equivalent (p>0.05). Rows with no superscripts were concluded equivalent (p<0.05)

Page 74: To my parents for their unyielding support and encouragement

74

Table 4-2. Continued

Syndrome Period 0.1 billion CFU 1 billion CFU 10 billion CFU Placebo

Epidermal Baseline 0.1 (± 0.1) 0.01 (± 0.01) 0.03 (± 0.01) 0.1 (± 0.02)

Week 1 0.02 (± 0.01) 0.04 (± 0.02) 0.04 (± 0.02) 0.1 (± 0.04)

Week 2 0.1 (± 0.1) 0.1 (± 0.04) 0.05 (± 0.03) 0.04 (± 0.03)

Week 3 0.02 (± 0.01) 0.1 (± 0.1) 0.04 (± 0.02) 0.1 (± 0.04)

Week 4 0.02 (± 0.01) 0.02 (± 0.02) 0.02 (± 0.02) 0.1 (± 0.04)

Washout 0.02 (± 0.02) 0.01 (± 0.01) 0.02 (± 0.02) 0.1 (± 0.04)

Ear-Nose-Throat Baseline 0.1 (± 0.4) 0.1 (± 0.04) 0.1 (± 0.04) 0.2 (± 0.1)

Week 1 0.4 (± 0.1) a 0.1 (± 0.03)

b 0.1 (± 0.04) 0.3 (± 0.2)

Week 2 0.2 (± 0.1) 0.2 (± 0.1) 0.2 (± 0.1) 0.3 (± 0.1)

Week 3 0.2 (± 0.1) 0.2 (± 0.1) 0.2 (± 0.1) 0.4 (± 0.2)

Week 4 0.2 (± 0.1) 0.2 (± 0.1) 0.2 (± 0.1) 0.3 (± 0.1)

Washout 0.2 (± 0.1) 0.2 (± 0.1) 0.1 (± 0.1) 0.2 (± 0.1)

Data presented as mean (± std. error). Intervention by week combinations not connected by the same letter were concluded as not equivalent (p>0.05). Rows with no superscripts were concluded equivalent (p<0.05).

Page 75: To my parents for their unyielding support and encouragement

75

Table 4-2. Continued

Syndrome Period 0.1 billion CFU 1 billion CFU 10 billion CFU Placebo

Behavioral Baseline 0.2 (± 0.1) a 0.2 (± 0.1)

b 0.5 (± 0.1)

c 0.5 (± 0.1)

c

Week 1 0.2 (± 0.1) a 0.1 (± 0.1)

a 0.6 (± 0.2)

b 0.4 (± 0.1)

b

Week 2 0.1 (± 0.1) a 0.1 (± 0.1)

a 0.5 (± 0.1)

b 0.4 (± 0.2)

b

Week 3 0.1 (± 0.1) a 0.2 (± 0.01)

ab 0.5 (± 0.1)

c 0.4 (± 0.2)

bc

Week 4 0.2 (± 0.1) a 0.1 (± 0.1)

a 0.5 (± 0.2)

b 0.6 (± 0.2)

b

Washout 0.3 (± 0.2) a 0.2 (± 0.1)

a 0.5 (± 0.2)

b 0.4 (± 0.1)

b

Emetic Baseline 0.003 (± 0.003) 0.1 (± 0.1) 0.1 (± 0.2) 0.05 (± 0.04)

Week 1 0.1 (± 0.04) 0.1 (± 0.04) 0.03 (± 0.01) 0.1 (± 0.1)

Week 2 0.1 (± 0.1) 0.1 (± 0.1) 0.1 (± 0.1) 0.1 (± 0.04)

Week 3 0.02 (± 0.02) 0.1 (± 0.1) 0.1 (± 0.04) 0.1 (± 0.1)

Week 4 0.1 (± 0.03) 0.1 (± 0.1) 0.03 (± 0.02) 0.1 (± 0.1)

Washout 0.1 (± 0.1) 0.1 (± 0.1) 0.08 (± 0.04) 0.1 (± 0.1)

Data presented as mean (± std. error). Intervention by week combinations not connected by the same letter were concluded as not equivalent (p>0.05). Rows with no superscripts were concluded equivalent (p<0.05).

Page 76: To my parents for their unyielding support and encouragement

76

Figure 4-2. Gastrointestinal distress syndrome

0

1

2

3

4

5

6

Baseline Week 1 Week 2 Week 3 Week 4 Washout

Mean S

yndro

me S

core

Gastrointestinal Distress

Placebo 0.1 billion 1 billion 10 billion

Page 77: To my parents for their unyielding support and encouragement

77

Figure 4-3. Cephalic syndrome

0

1

2

3

4

5

6

Baseline Week 1 Week 2 Week 3 Week 4 Washout

Mean S

yndro

me S

core

Cephalic

Placebo 0.1 billion 1 billion 10 billion

Page 78: To my parents for their unyielding support and encouragement

78

Figure 4-4. Epidermal syndrome

0

1

2

3

4

5

6

Baseline Week 1 Week 2 Week 3 Week 4 Washout

Mean S

yndro

me S

core

Epidermal

Placebo 0.1 billion 1 billion 10 billion

Page 79: To my parents for their unyielding support and encouragement

79

Figure 4-5. Ear nose and throat syndrome

0

1

2

3

4

5

6

Baseline Week 1 Week 2 Week 3 Week 4 Washout

Mean S

yndro

me S

core

Ear Nose and Throat

Placebo 0.1 billion 1 billion 10 billion

Page 80: To my parents for their unyielding support and encouragement

80

Figure 4-6. Behavioral syndrome

0

1

2

3

4

5

6

Baseline Week 1 Week 2 Week 3 Week 4 Washout

Mean S

yndro

me S

core

Behavrioal

Placebo 0.1 billion 1 billion 10 billion

Page 81: To my parents for their unyielding support and encouragement

81

Figure 4-7. Emetic syndrome

0

1

2

3

4

5

6

Baseline Week 1 Week 2 Week 3 Week 4 Washout

Mean S

yndro

me S

core

Emetic

Placebo 0.1 billion 1 billion 10 billion

Page 82: To my parents for their unyielding support and encouragement

82

Table 4-3. Daily Questionnaire symptom equivalence testing

Syndrome Period 0.1 billion CFU 1 billion CFU 10 billion CFU Placebo

Constipation Baseline 0.1 (± 0.1) a 0.4 (± 0.2) b 0.4 (± 0.2) c 0.2 (± 0.1) a

Week 1 0.2 (± 0.1) a 0.3 (± 0.1) b 0.7 (± 0.3) c 0.1 (± 0.04) a

Week 2 0.2 (± 0.01) a 0.3 (± 0.1) b 0.6 (± 0.2) c 0.3 (± 0.1) b

Week 3 0.1 (± 0.1) a 0.6 (± 0.2) ab 0.7 (± 0.3) b 0.3 (± 0.1) c

Week 4 0.1 (± 0.1) a 0.5 (± 0.1) b 0.7 (± 0.2) c 0.2 (± 0.1) a

Washout 0.1 (± 0.03) a 0.5 (± 0.2) b 0.5 (± 0.2) c 0.3 (± 0.1) bc

Diarrhea Baseline 0.1 (± 0.04) 0.2 (± 0.1) 0.2 (± 0.1) 0.2 (± 0.1)

Week 1 0.2 (± 0.03) 0.2 (± 0.1) 0.1 (± 0.03) 0.2 (± 0.1)

Week 2 0.1 (± 0.1) 0.3 (± 0.1) 0.3 (± 0.1) 0.2 (± 0.1)

Week 3 0.1 (± 0.1) a 0.4 (± 0.1)b 0.3 (± 0.1) a 0.2 (± 0.1) a

Week 4 0.2 (± 0.1) a 0.3 (± 0.1) b 0.1 (± 0.1) a 0.4 (± 0.1) b

Washout 0.1 (± 0.04) a 0.2 (± 0.1) b 0.2 (± 0.1) b 0.3 (± 0.1) b

Fatigue Baseline 0.7 (± 0.2) a 0.7 (± 0.2) a 0.8 (± 0.2) b 1.1 (± 0.3) b

Week 1 0.7 (± 0.2) a 0.5 (± 0.2) a 0.8 (± 0.2) b 0.9 (± 0.3) b

Week 2 0.6 (± 0.2) a 0.7 (± 0.2) b 0.9 (± 0.2) c 0.9 (± 0.3) d

Week 3 0.4 (± 0.1) a 0.6 (± 0.2) b 0.9 (± 0.3) c 1.0 (± 0.3) d

Week 4 0.5 (± 0.1) a 0.6 (± 0.2) ab 0.8 (± 0.2) c 0.8 (± 0.3) b

Washout 0.5 (± 0.2) a 0.5 (± 0.2) a 0.7 (± 0.2) c 0.6 (± 0.3) ac

Data presented as mean (± std. error). Intervention by week combinations not connected by the same letter were concluded as not equivalent (p>0.05). Rows with no superscripts were concluded equivalent (p<0.05).

Page 83: To my parents for their unyielding support and encouragement

83

Table 4-3. Continued

Syndrome Period 0.1 billion CFU 1 billion CFU 10 billion CFU Placebo

Satiety Baseline 0.7 (± 0.2) a 0.9 (± 0.2) a 0.9 (± 0.2) b 0.8 (± 0.2) b

Week 1 0.3 (± 0.1) a 0.8 (± 0.2) b 1.1 (± 0.3) c 0.7 (± 0.2) d

Week 2 0.3 (± 0.1) a 0.8 (± 0.2) b 0.9 (± 0.2) c 0.9 (± 0.3) c

Week 3 0.3 (± 0.1) a 0.8 (± 0.2) b 1.0 (± 0.3) b 0.9 (± 0.3) c

Week 4 0.4 (± 0.1) a 0.9 (± 0.3) b 0.9 (± 0.3) b 0.9 (± 0.3) c

Washout 0.3 (± 0.1) a 0.9 (± 0.3) b 0.8 (± 0.3) c 0.8 (± 0.3) c

Bowel Movements

Baseline 1.5 (± 0.2) ab 1.3 (± 0.1) a 1.5 (± 0.1) ab 1.55 (± 0.2) ab

Intervention (Weeks 1-4)

1.5 (± 0.1) ab 1.5 (± 0.1) ab 1.5 (± 0.1) ab 1.4 (± 0.1) a

Washout 1.4 (± 0.1) a 1.4 (± 0.1) b 1.6 (± 0.2) a 1.5 (± 0.2) c

Hours of Sleep

Baseline 6.6 (± 0.2) a 7.1 (± 0.1) b 6.6 (± 0.2) a 6.8 (± 0.2) c

Week 1 6.8 (± 0.2) a 7.1 (± 0.2) b 6.9 (± 0.2) c 6.7 (± 0.1) d

Week 2 6.8 (± 0.2) a 7.1 (± 0.2) b 6.7 (± 0.2) c 6.5 (± 0.2) d

Week 3 7.0 (± 0.2) a 7.3 (± 0.2) a 6.8 (± 0.2) b 6.6 (± 0.2) c

Week 4 7.0 (± 0.2) a 6.9 (± 0.2) a 6.9 (± 0.2) b 6.8 (± 0.2) c

Washout 7.0 (± 0.2) a 7.1 (± 0.1) b 6.7 (± 0.2) a 6.8 (± 0.2) c

Data presented as mean (± std. error). Intervention by week combinations not connected by the same letter were concluded as not equivalent (p>0.05).

Page 84: To my parents for their unyielding support and encouragement

84

Figure 4-8. Constipation symptom

0

1

2

3

4

5

6

Baseline Week 1 Week 2 Week 3 Week 4 Washout

Mean S

ym

pto

m S

core

Constipation

Placebo 0.1 billion 1 billion 10 billion

Page 85: To my parents for their unyielding support and encouragement

85

Figure 4-9. Diarrhea symptom

0

1

2

3

4

5

6

Baseline Week 1 Week 2 Week 3 Week 4 Washout

Mean S

ym

pto

m S

core

Diarrhea

Placebo 0.1 billion 1 billion 10 billion

Page 86: To my parents for their unyielding support and encouragement

86

Figure 4-10. Fatigue symptom

0

1

2

3

4

5

6

Baseline Week 1 Week 2 Week 3 Week 4 Washout

Mean S

ym

pto

m S

core

Fatigue

Placebo 0.1 billion 1 billion 10 billion

Page 87: To my parents for their unyielding support and encouragement

87

Figure 4-11. Bowel movement frequency

0

1

2

3

4

5

6

7

8

9

10

Baseline Intervention Washout

Mean B

ow

el M

ovem

ents

Bowel Movement Frequency

Placebo 0.1 billion 1 billion 10 billion

Page 88: To my parents for their unyielding support and encouragement

88

Figure 4-12. Hours of sleep

0

2

4

6

8

10

12

Baseline Week 1 Week 2 Week 3 Week 4 Washout

Mean H

ours

of S

leep

Hours of Sleep

Placebo 0.1 billion 1 billion 10 billion

Page 89: To my parents for their unyielding support and encouragement

89

Table 4-4. Gastrointestinal Symptom Rating Scale equivalence testing

Syndrome Period 0.1 billion CFU 1 billion CFU 10 billion CFU Placebo

Abdominal Pain Baseline 1.3 (± 0.1) 1.4 (± 0.1) 1.5 (± 0.2) 1.6 (± 0.1)

Intervention 1.5 (± 0.1) 1.6 (± 0.1) 1.6 (± 0.1) 1.8 (± 0.1)

Washout 1.6 (± 0.2) 1.6 (± 0.1) 1.3 (± 0.1) 1.7 (± 0.1)

Reflux Baseline 1.3 (± 0.2) 1.1 (± 0.1) 1.1 (± 0.1) 1.3 (± 0.1)

Intervention 1.3 (± 0.2) 1.1 (± 0.1) 1.2 (± 0.1) 1.3 (± 0.1)

Washout 1.3 (± 0.2) 1.1 (± 0.1) 1.2 (± 0.1) 1.3 (± 0.1)

Indigestion Baseline 1.4 (± 0.1) 1.5 (± 0.01) 1.7 (± 0.1) 1.8 (± 0.1)

Intervention 1.6 (± 0.1) 1.9 (± 0.1) 1.6 (± 0.1) 1.9 (± 0.2)

Washout 1.3 (± 0.1) 1.6 (± 0.1) 1.7 (± 0.1) 1.9 (± 0.1)

Constipation Baseline 1.1 (± 0.1) 1.4 (± 0.1) 1.4 (± 0.1) 1.5 (± 0.1)

Intervention 1.2 (± 0.1) 1.6 (± 0.2) 1.8 (± 0.2) 1.6 (± 0.1)

Washout 1.2 (± 0.1) 1.6 (± 0.2) 1.7 (± 0.2) 1.5 (± 0.1)

Diarrhea Baseline 1.2 (± 0.1) 1.3 (± 0.2) 1.4 (± 0.1) 1.3 (± 0.1)

Intervention 1.2 (± 0.1) 1.6 (± 0.2) 1.5 (± 0.1) 1.7 (± 0.2)

Washout 1.3 (± 0.1) 1.4 (± 0.2) 1.3 (± 0.1) 1.8 (± 0.2)

Data presented as mean (± std. error). Intervention by week combinations were all concluded equivalent (p<0.05).

Page 90: To my parents for their unyielding support and encouragement

90

Figure 4-13. GSRS abdominal pain syndrome

1

2

3

4

5

6

7

Baseline Intervention Washout

Mean S

yndro

me S

core

Abdominal Pain

Placebo 0.1 billion 1 billion 10 billion

Page 91: To my parents for their unyielding support and encouragement

91

Figure 4-14. GSRS reflux syndrome

1

2

3

4

5

6

7

Baseline Intervention Washout

Mean S

yndro

me S

core

Reflux

Placebo 0.1 billion 1 billion 10 billion

Page 92: To my parents for their unyielding support and encouragement

92

Figure 4-15. GSRS indigestion syndrome

1

2

3

4

5

6

7

Baseline Intervention Washout

Mean S

yndro

me S

core

Indigestion

Placebo 0.1 billion 1 billion 10 billion

Page 93: To my parents for their unyielding support and encouragement

93

Figure 4-16. GSRS constipation syndrome

1

2

3

4

5

6

7

Baseline Intervention Washout

Mean C

onstipation S

core

Constipation

Placebo 0.1 billion 1 billion 10 billion

Page 94: To my parents for their unyielding support and encouragement

94

Figure 4-17. GSRS diarrhea syndrome

1

2

3

4

5

6

7

Baseline Intervention Washout

Mean S

yndro

me S

core

Diarrhea

Placebo 0.1 billion 1 billion 10 billion

Page 95: To my parents for their unyielding support and encouragement

95

Table 4-5. Gastrointestinal viability of Bacillus spores

Period Placebo 0.1 billion CFU 1 billion CFU 10 billion CFU

log CFU/g Baseline 0.9 ± 0.1 0.9 ± 0.1 0.9 ± 0.1 1.2 ± 0.2

Intervention 1.1 ± 0.1b 4.6 ± 0.1a 5.6 ± 0.1a 6.4 ± 0.1a

Washout 1.6 ± 0.1ab 1.3 ± 0.2ab 0.8 ± 0.1a 2.1 ± 0.1b

Data presented as mean (± std. error). Intervention by week combinations were all concluded equivalent (p<0.05).

Figure 4-18. Gastrointestinal viability presented as means ± SEM

0

1

2

3

4

5

6

7

Baseline Intervention Washout

Log10 C

FU

/G

Gastrointestinal Viability

Placebo 0.1 billion 1 billion 10 billion

Page 96: To my parents for their unyielding support and encouragement

96

CHAPTER 5 DISCUSSION AND CONCLUSIONS

Discussion

The spore forming bacteria Bacillus has long been thought to live its life cycle in

the soil, however recovery of vegetative cells from soil samples has been challenging

(71). Fecal samples collected from healthy adults consuming a western diet typically

contain about 104 spores of Bacillus / g of feces. This data suggests that Bacillus

species are natural residents of the human GIT and their residence in soil only serves

as a reservoir for the spore form (62). The use of Bacillus species as probiotics has also

been demonstrated in several studies demonstrating their ability to act as competitive

exclusion agents (48, 72), production of antimicrobials (72), and immunostimulatory

properties (73). All of these properties have been demonstrated in B. subtilis though to

date a clinical evaluation of B. subtilis on general wellness and gastrointestinal health as

a single probiotic agent has not been conducted. The continued use of Bacillus species

in foods and as supplements requires its evaluation at typical therapeutic dosages (i.e.

greater than 0.1 billion CFU/capsule/day). It is with this in mind that a double-blind,

placebo-controlled, randomized, clinical trial (n=81) was conducted to evaluate the

effects of Bacillus subtilis R0179 consumption on general wellness, gastrointestinal

function and gastrointestinal viability in healthy adults. Participants were randomized to

one of four treatment groups (0.1, 1, 10 billion CFU/capsule/day or placebo) for a period

of four weeks and general wellness as well as gastrointestinal symptoms were

evaluated using questionnaires. Stool samples were collected to assess the

gastrointestinal transit viability and impact on the intestinal microbiome of the B. subtilis

R0179.

Page 97: To my parents for their unyielding support and encouragement

97

Throughout this six week clinical trial, two participants reported adverse effects

while consuming the probiotic. One participant in the 0.1 billion CFU treatment group

experienced diarrheal symptoms and completed the study, this event did not occur in

any other participants and did not persist. A participant in the 10 billion CFU treatment

group experienced itchiness and redness symptoms that were sporadic and persisted

for several days. The participant voluntarily withdrew from the study, these symptoms

did not occur in any other participants. A third participant reported a serious adverse

event of hospitalization with an admitting diagnosis of hypotension, but did not

discontinue probiotic consumption and completed the study. Despite these adverse

effects, the probiotics were well tolerated by a majority of the study participants.

Questionnaire data on general wellness and gastrointestinal symptom data was

evaluated using equivalence testing. The validated GSRS questionnaire was used to

evaluate gastrointestinal symptoms (42). Mean scores for the abdominal pain, reflux,

indigestion, constipation and diarrhea syndromes for each treatment group (0.1, 1, 10

billion CFU/capsule/day and placebo) across each treatment period (baseline,

treatment, washout) were all below the clinically significant score of 2 (slight discomfort)

on the GSRS. Equivalence testing revealed that all treatment group means across each

treatment period were concluded equivalent.

Daily questionnaires were used to evaluate general wellness in study

participants. Mean scores for the GI distress, cephalic, ENT, behavioral, emetic, and

epidermal syndromes for each treatment group across each week of the study were all

below the clinically significant score of on the DQ. The ungrouped symptoms,

constipation and diarrhea were also below the clinically significant score of 1 on the DQ.

Page 98: To my parents for their unyielding support and encouragement

98

The only symptom that exceeded the clinical threshold was the fatigue symptom by the

placebo group during the baseline week and week 3 of the intervention period.

Evaluations of mean hours of sleep and mean bowel movement frequency revealed that

participants were only getting between 6.5 and 7.3 hours of sleep per night and were

averaging 1.4 to 1.6 bowel movements per day. Participants in the placebo group who

were getting the least amount of sleep also reported higher fatigue scores. Conversely,

all treatment groups averaged more than one bowel movement per day which is further

confirmed by their low constipation and diarrhea symptom scores.

Equivalence testing of syndrome and symptom scores revealed a number of

treatment group and period combinations that were concluded were not equivalent,

however no clear trends could be identified. This is particularly clear when intervention

groups were concluded not equivalent in the baseline week and this non-equivalence

continued well into the intervention and washout periods. Kruskall-Wallis tests

confirmed that intervention groups did not change from their baseline values.

Furthermore, when conducting these statistical analyses a very conservative

predetermined practical difference threshold of one unit on the questionnaire was used

in order to ensure non-equivalence could be detected. If that pre-determined practical

difference threshold is tested again at two units on the DQ, then all previously non-

equivalent results are concluded as equivalent.

This use of equivalence testing on symptom data is the first of its kind to be used

in a clinical trial. Standards of care are continually being redefined due to advances in

medicine. Measuring the efficacy of novel therapies against traditional therapies is one

method used to encourage new standards of care. These comparative studies evaluate

Page 99: To my parents for their unyielding support and encouragement

99

the superiority of one therapy over another. The opposite is true of equivalence tests,

also referred to as two-one sided tests (TOST). When testing for equivalence, such as

when comparing multiple dosages, traditional hypotheses testing methods are reversed.

A non-significant value, of a traditional comparative test, does not prove that two

treatments are equivalent, it only proves that the treatments may be equivalent although

equivalence is usually not well-defined in superiority testing. For example, a -3%

difference in the eradication rate of H. pylori infection, when comparing a new therapy to

a traditional therapy, may not be significantly different but this does not mean that they

are equivalent, the new therapy is less effective and thus should not be recommended

to patients (74).

A study conducted by Barker et al.2002 evaluated the results of traditional two-

sided tests to that of the TOST using National Immunization Survey data of the number

of vaccinations that while children used when compared to different ethnicities. Nine out

of twenty-one comparisons were contradictory when using the different comparison

methods (75). This disparity is further exacerbated by the fact that superiority testing

gives no indication of how similar differing treatments are only that one is superior or

that it cannot be proven to be superior with the current set of data. The TOST provides

a method by which the similarity of treatment groups can be compared according to a

pre-specified clinically relevant difference threshold. It is for this reason that we set our

practical difference threshold to one-unit when conducting equivalence tests.

This method has demonstrated that there were some non-equivalent values

when comparing mean syndrome and symptom scores, but the scores were no different

from their base line values. Additionally, syndrome and symptom means were below

Page 100: To my parents for their unyielding support and encouragement

100

clinically-different threshold values and so, consumption of B. subtilis R0179 does not

have a negative impact on gastrointestinal health and general wellness symptoms at

dosages up to 10 billion CFU/capsule/day. While the safety of B. subtilis R0179 has

already been demonstrated by over 100 clinical trials as reviewed in Tompkins et al.

2010, Chen et al. 2012, Liu et al. 2013, and Hu et al. 2013 until now the effect of R0179

consumption on gastrointestinal and general wellness has never been fully evaluated

especially at dosages higher than 0.1 billion CFU/day.

Analysis of the viability and recovery of the ingested spores from fecal samples

indicated a clear dose response among the treatment groups. Each group was

significantly different from one another with the most spores being recovered from the

10 billion CFU/capsule/day treatment group and the least being recovered from the

placebo group. Furthermore, our results complement data reported by Hong et al. 2009

and Tompkins et al. 2010 which indicate that Bacillus spores transiently colonize the

human gastrointestinal tract and have low adhesive potential

Conclusions

This is the first assessment of Bacillus subtilis R0179 as a single probiotic agent

in a double-blind, placebo-controlled, randomized, clinical trial. Consumption of the

bacterial strain for a period of six weeks did not produce any persisting adverse effects.

Recovery of the probiotic in fecal samples demonstrated a dose response relationship,

and leads further credence to the transitory nature of the bacteria in the human

gastrointestinal tract. The results of this study indicate that the regular consumption of

the B. subtilis R0179 is safe and well tolerated in healthy adults even at high dosages

not typically found in probiotic supplements.

Page 101: To my parents for their unyielding support and encouragement

101

APPENDIX A IRB APPROVAL

Page 102: To my parents for their unyielding support and encouragement

102

APPENDIX B INFORMED CONSENT

INFORMED CONSENT FORM to Participate in Research, and

AUTHORIZATION to Collect, Use, and Disclose Protected

Health Information (PHI)

INTRODUCTION

Name of person seeking your consent: Place of employment & position: Please read this form which describes the study in some detail. A member of the research team will describe this study to you and answer all of your questions. Your participation is entirely voluntary. If you choose to participate you can change your mind at any time and withdraw from the study. You will not be penalized in any way or lose any benefits to which you would otherwise be entitled if you choose not to participate in this study or to withdraw. If you have questions about your rights as a research subject, please call the University of Florida Institutional Review Board (IRB) office at (352) 273-9600.

GENERAL INFORMATION ABOUT THIS STUDY

1. Name of Participant ("Study Subject") ___________________________________________________________________

2. What is the Title of this research study?

Evaluation of Bacillus subtilis R0179 in capsules on gastrointestinal survival, gastrointestinal symptoms and general wellness in healthy young adults.

Page 103: To my parents for their unyielding support and encouragement

103

3. Who do you call if you have questions about this research study?

Principal Investigator: Dr. Wendy Dahl Work: 352-392-1991 ext. 224 Cell: 352-226-1773 Home: 352-374-7798 email: [email protected]

4. Who is paying for this research study?

The sponsor of this study is Institut Rosell Inc.

5. Why is this research study being done?

The purpose of this research study is to determine the survival of three doses of the probiotic B. subtilis (Bacillus subtilis R0179) delivered in capsules on survival through the gastrointestinal tract, impact on intestinal microbes, gastrointestinal symptoms and general well-being in healthy adults age 18 to 50.

You are being asked to be in this research study because you are a healthy adult between the age of 18 and 50 and meet the inclusion and exclusion criteria.

WHAT CAN YOU EXPECT IF YOU PARTICIPATE IN THIS STUDY?

6. What will be done as part of your normal clinical care (even if you did not participate in this research study)?

This study is not related to your normal clinical care.

7. What will be done only because you are in this research study? If you decide to take part in this study, following informed consent, you will begin a 7 day baseline period completing a daily questionnaire (paper or electronic) about bowel movement frequency and gastrointestinal symptoms. The questions will ask you about your bowel movements and gastrointestinal discomfort. Next, you will be randomized to receive the probiotic B. subtilis or placebo and begin taking one capsule per day for 28 days. At the randomization appointment you will be asked to complete the daily questionnaire, global physical activity questionnaire and the gastrointestinal symptom response scale, and height and weight will be determined. You will also continue the daily questionnaire for the 28 days of treatment. If you decide to take part in this study, you will be randomly assigned (much like the flip of a coin) to receive either [study substance] or placebo. A placebo is a substance that looks like and is given in the same way as an experimental treatment but contains no medicine, for example [a sugar pill, an injection of saline (salt water)]. A placebo is used in research studies to show what effect a treatment has compared with taking nothing at

Page 104: To my parents for their unyielding support and encouragement

104

all. If you are assigned to receive placebo, you will not receive the benefits of the [study substance], if there are any, nor will you be exposed to its risks, which are described below under "What are the possible discomforts and risks?" Studies have shown, however, that about 1 in 3 persons who take a placebo do improve, if only for a short time. You and the physician and other persons doing the study will not know whether you are receiving placebo or [study substance], but that information is available if it is needed. Also, you will have a 50% chance of receiving [study substance] and a 50% chance of receiving placebo. In the remainder of the description of what will be done, both the [study substance] and the placebo will be called "study treatment."

You will be randomized to one of the following groups:

Group 1: B. subtilis R0179 in capsules (approximately 10 billion colony forming units/serving) for period of 28 days.

Group 2: B. subtilis R0179 in capsules (approximately 1 billion colony forming units/serving), for a period of 28 days.

Group 3: B. subtilis R0179 in capsules (approximately 0.1 billion colony forming units/serving), for a period of 28 days.

Group 4: Placebo capsules for a period of 28 days.

In addition, consented participants in each group will be asked to collect one fecal sample at baseline, one fecal sample in week 4 and an additional sample after 7 days following the treatment and drop off to the Food Science and Human Nutrition building within 4 hours of collection. Participants will be provided with specialized plastic containers that can be placed over the toilet seat for fecal collection at home or in the clinical lab restroom facility.

In addition to fecal sample drop off dates, you will be expected to come to the clinical lab in the Food Science and Human Nutrition building for a final visit. At the final appointment you will have your weight measured and you will complete the general wellness questionnaire, GPAQ and GSRS again. Once the intervention period is over, you will be asked to complete a final GSRS on or about 7 days post treatment and continue with the daily questionnaire for 7 days post treatment.

If you have any questions now or at any time during the study, please contact one of the research team members listed in question 3 of this form.

8. How long will you be in this research study?

The total amount of time you will spend participating in the study is 42 days.

Page 105: To my parents for their unyielding support and encouragement

105

9. How many people are expected to take part in this research study?

Eighty people will be participating in this research study.

WHAT ARE THE RISKS AND BENEFITS OF THIS STUDY AND WHAT ARE YOUR OPTIONS?

10. What are the possible discomforts and risks from taking part in this research study?

Some people may feel uncomfortable when body weight is measured.

Some study participants may feel uncomfortable answering questions about their stool habits.

Some study participants may feel uncomfortable collecting stool samples.

Participation in more than one research study or project may cause risks to you. If you are already enrolled in another research study, please inform Dr. Wendy Dahl listed in question 3 of this consent form) or the person reviewing this consent with you before enrolling in this or any other research study or project.

Throughout the study, the researchers will notify you of new information that may become available and might affect your decision to remain in the study.

If you wish to discuss the information above or any discomforts you may experience, please ask questions now or call the PI or contact person listed on the front page of this form.

Other possible risks to you may include: none

Researchers will take appropriate steps to protect any information they collect about you. However, there is a slight risk that information about you could be revealed inappropriately or accidentally. Depending on the nature of the information, such a release could upset or embarrass you, or possibly affect your insurability or employability. Questions 17-21 in this form discuss what information about you will be collected, used, protected, and shared.

This study may include risks that are unknown at this time.

Participation in more than one research study or project may further increase the risks to you. If you are already enrolled in another research study, please inform one of the research team members listed in question 3 of this form or the person reviewing this consent with you before enrolling in this or any other research study or project.

Throughout the study, the researchers will notify you of new information that may become available and might affect your decision to remain in the study.

Page 106: To my parents for their unyielding support and encouragement

106

If you wish to discuss the information above or any discomforts you may experience, please ask questions now or call one of the research team members listed in question 3 in this form.

11a. What are the potential benefits to you for taking part in this research study?

There is no direct benefit to you for participating in this research study.

11b. How could others possibly benefit from this study?

Research into the efficacy of probiotic survival may lead to a better understanding of B. subtilis as a single probiotic agent.

11c. How could the researchers benefit from this study?

In general, presenting research results helps the career of a scientist. Therefore, the Principal Investigator listed in question 3 of this form may benefit if the results of this study are presented at scientific meetings or in scientific journals.

12. What other choices do you have if you do not want to be in this study?

You have the option to not take part in this study. If you do not want to take part in this study tell the Principle Investigator and do not sign this consent form.

13a. Can you withdraw from this study?

You are free to withdraw your consent and to stop participating in this study at any time. If you do withdraw your consent, you will not be penalized in any way and you will not lose any benefits to which you are entitled.

If you decide to withdraw your consent to participate in this study for any reason, please contact one of the research team members listed in question 3 of this form. They will tell you how to stop your participation safely.

If you have any questions regarding your rights as a research subject, please call the Institutional Review Board (IRB) office at (352) 273-9600.

13b. If you withdraw, can information about you still be used and/or collected?

If you withdraw your consent, your information will not be used.

13c. Can the Principal Investigator withdraw you from this study?

You may be withdrawn from the study without your consent for the following reasons:

You did not follow the instructions given, e.g. did not consume the probiotics or complete the daily diary.

Page 107: To my parents for their unyielding support and encouragement

107

WHAT ARE THE FINANCIAL ISSUES IF YOU PARTICIPATE?

14. If you choose to take part in this research study, will it cost you anything? The sponsor will only pay for medical services that you receive as part of your participation in this study as described in question 7 above. All other medical services that you would have received if you were not in this study will be billed to you or your insurance company. You will be responsible for paying any deductible, co-insurance, co-payments for those services and for any non-covered or out –of –network services. Some insurance companies may not cover costs associated with studies. Please contact your insurance company for additional information. The probiotic B. subtilis, questionnaires, and sample containers will be provided at no cost to you while you are participating in this study. The Sponsor will pay for medical services or activities required as part of your participation in this study. There will be no cost to you. If you receive a bill related to this study, please contact Wendy Dahl, 352 392 1991 ext 224. All other medical services provided to you that are not directly related to the study will be billed to you or your insurance company. You will be responsible for paying any deductible, co-insurance, and/or co-payments for these services, and any non-covered or out-of-network services.

Some insurance companies may not cover costs associated with studies. Please contact your insurance company for additional information.

15. Will you be paid for taking part in this study?

Yes, you will be paid $15 per stool and $155 for taking the capsules and completing the questionnaires for a maximum total of $200 for participating in the entire study. If you are paid for taking part in this study, your name and social security number will be reported to the appropriate University employees for purposes of making and recording the payment. You are responsible for paying income taxes on any payments provided by the study. If the payments total $600 or more, the University must report the amount you received to the Internal Revenue Service (IRS).

16. What if you are injured because of the study?

If you are injured as a direct result of your participation in this study, only the professional services that you receive from any University of Florida Health Science Center healthcare provider will be provided without charge. These healthcare providers include physicians,

Page 108: To my parents for their unyielding support and encouragement

108

physician assistants, nurse practitioners, dentists or psychologists. Any other expenses, including Shands hospital expenses, will be billed to you or your insurance provider.

You will be responsible for any deductible, co-insurance, or co-payments. Some insurance companies may not cover costs associated with research studies or research-related injuries. Please contact your insurance company for additional information.

Please contact one of the research team members listed in question 3 of this form if you experience an injury or have questions about any discomforts that you experience while participating in this study.

17. How will your health information be collected, used and shared?

If you agree to participate in this study, the Principal Investigator will create, collect, and use private information about you and your health. This information is called protected health information or PHI. In order to do this, the Principal Investigator needs your authorization The following section describes what PHI will be collected, used and shared, how it will be collected, used, and shared, who will collect, use or share it, who will have access to it, how it will be secured, and what your rights are to revoke this authorization.

Your protected health information may be collected, used, and shared with others to determine if you can participate in the study, and then as part of your participation in the study. This information can be gathered from you or your past, current or future health records, from procedures such as physical examinations, x-rays, blood or urine tests or from other procedures or tests. This information will be created by receiving study treatments or participating in study procedures, or from your study visits and telephone calls. More specifically, the following information may be collected, used, and shared with others:

Name

Address

Phone Number

Your social security number for compensation purposes

Body weight

Height

Age

Sex

Gastrointestinal wellness and symptoms

Stool microbiota data

This information will be stored in locked filing cabinets or on computer servers with secure passwords, or encrypted electronic storage devices.

Some of the information collected could be included in a "limited data set" to be used for other research purposes. If so, the limited data set will only include information that does not directly identify you. For example, the limited data set cannot include your name, address, telephone number, social security number, photographs, or other codes that link you to the information in the limited data set. If limited data sets are created and used,

Page 109: To my parents for their unyielding support and encouragement

109

agreements between the parties creating and receiving the limited data set are required in order to protect your identity and confidentiality and privacy.

18. For what study-related purposes will your protected health information be collected, used, and shared with others?

Your PHI may be collected, used, and shared with others to make sure you can participate in the research, through your participation in the research, and to evaluate the results of the research study. More specifically, your PHI may be collected, used, and shared with others for the following study-related purpose(s): To determine the survival of three doses of B. subtilis delivered in capsules on transit survival, impact on the intestinal microbiome, gastrointestinal symptoms and general well-being in healthy human adults age 18 to 50.

Once this information is collected, it becomes part of the research record for this study.

19. Who will be allowed to collect, use, and share your protected health information?

Only certain people have the legal right to collect, use and share your research records, and they will protect the privacy and security of these records to the extent the law allows. These people include:

The study Principal Investigator (listed in question 3 of this form) and research staff associated with this project.

Other professionals at the University of Florida or Shands Hospital that provide study-related treatment or procedures.

The University of Florida Institutional Review Board (IRB; an IRB is a group of people who are responsible for looking after the rights and welfare of people taking part in research).

20. Once collected or used, who may your protected health information be shared with?

Your PHI may be shared with:

The study sponsor (listed in Question 4 of this form).

United States and foreign governmental agencies who are responsible for overseeing research, such as the Food and Drug Administration, the Department of Health and Human Services, and the Office of Human Research Protections .

Government agencies who are responsible for overseeing public health concerns such as the Centers for Disease Control and federal, state and local health departments.

Otherwise, your research records will not be released without your permission unless required by law or a court order. It is possible that once this information is shared with

Page 110: To my parents for their unyielding support and encouragement

110

authorized persons, it could be shared by the persons or agencies who receive it and it would no longer be protected by the federal medical privacy law.

21. If you agree to take part in this research study, how long will your protected health information be used and shared with others?

Your PHI will be used and shared with others for one year following the completion of the study.

You are not required to sign this consent and authorization or allow researchers to collect, use and share your PHI. Your refusal to sign will not affect your treatment, payment, enrollment, or eligibility for any benefits outside this research study. However, you cannot participate in this research unless you allow the collection, use and sharing of your protected health information by signing this consent and authorization.

You have the right to review and copy your protected health information. However, we can make this available only after the study is finished.

You can revoke your authorization at any time before, during, or after your participation in this study. If you revoke it, no new information will be collected about you. However, information that was already collected may still be used and shared with others if the researchers have relied on it to complete the research. You can revoke your authorization by giving a written request with your signature on it to the Principal Investigator.

SIGNATURES

As an investigator or the investigator’s representative, I have explained to the participant the purpose, the procedures, the possible benefits, and the risks of this research study; the alternative to being in the study; and how the participant’s protected health information will be collected, used, and shared with others:

Signature of Person Obtaining Consent and Authorization

Date

Page 111: To my parents for their unyielding support and encouragement

111

You have been informed about this study’s purpose, procedures, possible benefits, and risks; the alternatives to being in the study; and how your protected health information will be collected, used and shared with others. You have received a copy of this Form. You have been given the opportunity to ask questions before you sign, and you have been told that you can ask questions at any time. You voluntarily agree to participate in this study. You hereby authorize the collection, use and sharing of your protected health information as described in sections 17-21 above. By signing this form, you are not waiving any of your legal rights.

Signature of Person Consenting and Authorizing Date

Page 112: To my parents for their unyielding support and encouragement

112

APPENDIX C GLOBAL PHYSICAL ACTIVITY QUESTIONNNAIRE

Page 113: To my parents for their unyielding support and encouragement

113

Page 114: To my parents for their unyielding support and encouragement

114

Page 115: To my parents for their unyielding support and encouragement

115

APPENDIX D DAILY QUESTIONNAIRE

Did you experience any of the following symptoms in the last 24 hours? Please circle one choice 0-10, 0= none, 3= mild, 6= very severe. Note: If you are a female, please do not rate menstrual cramping and bloating. Please rate the severity by circling the appropriate number below Bloating 0 1 2 3 4 5 6 None moderate very severe Flatulence 0 1 2 3 4 5 6 None moderate very severe Abdominal Cramping 0 1 2 3 4 5 6 None moderate very severe Stomach Noises 0 1 2 3 4 5 6 None moderate very severe Headache 0 1 2 3 4 5 6 None moderate very severe Dizziness

0 1 2 3 4 5 6 None moderate very severe Sore throat 0 1 2 3 4 5 6 None moderate very severe

Page 116: To my parents for their unyielding support and encouragement

116

Blocked ear canal 0 1 2 3 4 5 6 None moderate very severe Nasal congestion

0 1 2 3 4 5 6 None moderate very severe Runny eyes 0 1 2 3 4 5 6 None moderate very severe Nausea

0 1 2 3 4 5 6 None moderate very severe Vomiting

0 1 2 3 4 5 6 None moderate very severe Diarrhea

0 1 2 3 4 5 6 None moderate very severe Feeling of constipation

0 1 2 3 4 5 6 None moderate very severe Feeling of satiety (not hungry)

0 1 2 3 4 5 6 None moderate very severe Itching

0 1 2 3 4 5 6 None moderate very severe

Page 117: To my parents for their unyielding support and encouragement

117

Fatigue 0 1 2 3 4 5 6 None moderate very severe Skin rash

0 1 2 3 4 5 6 None moderate very severe Skin redness/flushing

0 1 2 3 4 5 6 None moderate very severe Feelings of anxiety

0 1 2 3 4 5 6 None moderate very severe Feelings of depression 0 1 2 3 4 5 6 None moderate very severe How many stools (bowel movements) did you have today? ______________

0 1 2 3 4 5 6 7 8 9 10 >10 What is your energy level today?

Excellent Very good Good Fair Poor

How many hours did you sleep last night? Do not include the time it took you to fall

asleep or anytime you were awakened during the night. Circle the time that most closely matches your sleep.

<5 hours 5-6 hours 6-7 hours 7-8 hours 8-9 hours >9 hours

Did you take your probiotic capsule today? Yes No Did you visit a doctor today? Yes No

Did you take an antibiotic today? Yes No

Page 118: To my parents for their unyielding support and encouragement

118

APPENDIX E GASTROINTESTINAL SYMPTOM RESPONSE SCALE

(1) No discomfort at all (5) Moderately severe discomfort (2) Slight discomfort (6) Severe discomfort (3) Mild discomfort (7) Very severe discomfort (4) Moderate discomfort Questions: Answer each question using the response scale above.

_____Have you been bothered by stomach ache or pain during the past week?

(Stomach ache refers to all kinds of aches or pains in your stomach or belly) _____Have you been bothered by heartburn during the past week? (By heartburn we mean a burning pain or discomfort behind the breastbone in your chest) _____Have you been bothered by acid reflux during the past week? (By acid reflux we mean regurgitation or flow of sour or bitter fluid into your mouth) _____Have you been bothered by hunger pains in the stomach or belly during the

past week? (This hallow feeling in the stomach is associated with the need to eat between meals.) _____Have you been bothered by nausea during the past week?

(By nausea we mean a feeling of wanting to be sick) _____Have you been bothered by rumbling in your stomach or belly during the past week? (Rumbling refers to vibrations or noises in the stomach) _____Has your stomach felt boated during the past week? (Feeling bloated refers to swelling in the stomach or belly) _____Have you been bothered by burping during the past week?

(Burping refers to bringing up air or gas through the mouth) _____Have you been bothered by passing gas or flatus during the past week? (Passing gas refers to the release of air or gas from the bowels) _____Have you been bothered by constipation during the past week?

(Constipation refers to a reduced ability to empty the bowels) _____Have you been bothered by diarrhea during the past week? (Diarrhea refers to frequent loose or watery stools) _____Have you been bothered by loose stools during the past week?

Page 119: To my parents for their unyielding support and encouragement

119

(If your stools have been alternately hard and loose, this question only refers to the extent you have been bothered by the stools being loose) _____Have you been bothered by hard stools during the past week? (If your stools have been alternately hard and loose, this question only refers to the extent you have been bothered by stools being hard) _____Have you been bothered by an urgent need to have a bowel movement during the past week? (This urgent need to open your bowels makes you rush to the toilet) _____When going to the toilet during the past week, have you had the feeling of not

completely emptying your bowels? (The feeling that after finishing a bowel

movement, there is still some stool that needs to be passed)

Page 120: To my parents for their unyielding support and encouragement

120

LIST OF REFERENCES

1. Thum C, Cookson AL, Otter DE, McNabb WC, Hodgkinson AJ, Dyer J, Roy NC. Can nutritional modulation of maternal intestinal microbiota influence the development of the infant gastrointestinal tract? J Nutr. 2012;142:1921-8.

2. Salminen S, Gibson GR, McCartney AL, Isolauri E. Influence of mode of delivery on gut microbiota composition in seven year old children. Gut An international Journal of Gastroenterology and Hepatology. 2004;53:1388-1389.

3. Mshvildadze M, Mai V. Intestinal microbiota development in the premature neonate: establishment of a lasting commensal relationship? Nutrition Reviews. 2008;66:658-63.

4. Grölund M-M, Lehtonen O-P, Eerola E, Kero P. Fecal Microflora in Healthy Infants Born by Different Methods of Delivery: Permanent Changes in Intestinal Flora After Cesarean Delivery. Journal of Pediatric Gastroenterology & Nutrition. 1999;28:19-25.

5. Hopkins MJ, Sharp R, Macfarlane GT. Age and disease related changes in intestinal bacterial populations assessed by cell culture, 16S rRNA abundance, and community cellular fatty acid profiles. Gut An international Journal of Gastroenterology and Hepatology. 2001;48:198-205.

6. Biagi E, Bologna Uo, Candela M, Bologna Uo, Fairweather-Tait S, Anglia UoE, Franceschi C, Bologna Uo, Brigidi P, et al. Ageing of the human metaorganism: the microbial counterpart. AGE. 2013;34:247-67.

7. Noverr MC, Huffnagle GB. Does the microbiota regulate immune responses outside the gut? Trends in Microbiology. 2004;12:562-8.

8. Bäckhed F, Fraser CM, Ringel Y, Sanders ME, Sartor BR, Sherman PM, Versalovic J, Young V, Finlay BB. Defining a Healthy Human Gut Microbiome: Current Concepts, Future Directions, and Clinical Applications. Cell Host & Microbe. 2012;12:611-22.

9. Curtis Huttenhower THMPC. Structure, function and diversity of the healthy human microbiome. Nature. 2012;486:207-14.

10. Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, Sogin ML, Jones WJ, Roe BA, et al. A core gut microbiome in obese and lean twins. Nature. 2009;457:480-4.

11. Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, Nielsen T, Pons N, Levenez F, et al. A human gut microbial gene catalog established by metagenomic sequencing. Nature. 2010;464:59-65.

Page 121: To my parents for their unyielding support and encouragement

121

12. Jernberg C, L S, [ouml], fmark, Edlund C, Jansson JK. Long-term ecological impacts of antibiotic administration on the human intestinal microbiota. The ISME Journal. 2007;1:56-66.

13. Alonso VR, Guarnera, Francisco. Linking the gut microbiota to human health. British Journal of Nutrition. 2013, 2013;109.

14. Metchnikoff E, Chalmers MP. The prolongation of life; optimistic studies : Metchnikoff, Elie, 1845-1916 Internet Archive. New York & London: G.P. Putnam's Sons; 1907.

15. Sekirov I, Russell SL, Antunes LCM, Finlay BB. Gut Microbiota in Health and Disease. 2010;90:859-904.

16. FAO/WHO. Guidelines for the Evaluation of Probiotics in Food. Report of a Joint FAO/WHO Working Group on Drafting Guidelines for the Evaluation of Probiotics in Food. London, England and Ontario, Canada; 2002.

17. Salminen S, Wright Av, Ouwehand A. Safety assessment of starters and probiotics. In: Adams M, Nout R, editors. Fermentation and Food Safety. Gaithersburg, MD: Aspen Publishers; 2000. p. 239-52.

18. Holfzapfel WH. Introduction to Prebiotics and Probiotics. In: Goktepe I, Juneja VK, Ahmedna M, editors. Probiotics in Food Safety and Human Health. Boca Raton, FL: Taylor & Franics Group; 2006. p. 1-25.

19. Saarela M, Mogensen G, Fonden R, Matto J, Mattila-Sandholm T. Probiotic bacteria: safety, functional and technological properties. J Biotechnol. 2000;84:197-215.

20. Vaughan EE, Mollet B. Probiotics in the new millennium. Molecular Nutrition. 1999;43:148-53.

21. Fooks LJ, Gibson GR. Probiotics as modulators of the gut flora. British Journal of Nutrition. 2002;88:39-49.

22. Mercenier A, Pavan S, Pot B. Probiotics as biotherapeutic agents: present knowledge and future prospects. Curr Pharm Des. 2003;9:175-91.

23. Supplements FooD. Q&A on Dietary Supplements. 2013 [cited 4/15/2013]; At time of access: Last updated 4/11/13:[Available from:http://www.fda.gov/Food/DietarySupplements/QADietarySupplements]

24. Marteau P. Safety aspects of probiotic products. Scandinavian Journal of Nutrition. 2001;45:22-4.

25. Hennequin C, Kauffmann-Lacroix C, Jobert A, Viard JP, Ricour C, Jacquemin JL, Berche P. Possible role of catheters in Saccharomyces boulardii fungemia. Eur J Clin Microbiol Infect Dis. 2000 Jan;19:16-20.

Page 122: To my parents for their unyielding support and encouragement

122

26. Bleichner G, Blehaut H, Mentec H, Moyse D. Saccharomyces boulardii prevents diarrhea in critically ill tube-fed patients. A multicenter, randomized, double-blind placebo-controlled trial. Intensive Care Med. 1997;23:517-23.

27. Saxelin M, Chuang NH, Chassy B, Rautelin H, Makela PH, Salminen S, Gorbach SL. Lactobacilli and bacteremia in southern Finland, 1989-1992. Clin Infect Dis. 1996 ;22:564-6.

28. Salminen MK, Tynkkynen S, Rautelin H, Saxelin M, Vaara M, Ruutu P, Sarna S, Valtonen V, Jarvinen A. Lactobacillus bacteremia during a rapid increase in probiotic use of Lactobacillus rhamnosus GG in Finland. Clin Infect Dis. 2002;35:1155-60.

29. Dussurget O, Cabanes D, Dehoux P, Lecuit M, Buchrieser C, Glaser P, Cossart P. Listeria monocytogenes bile salt hydrolase is a PrfA-regulated virulence factor involved in the intestinal and hepatic phases of listeriosis. Mol Microbiol. 2002;45:1095-106.

30. Kandell RL, Bernstein C. Bile salt/acid induction of DNA damage in bacterial and mammalian cells: implications for colon cancer. Nutr Cancer. 1991;16:227-38.

31. Franz CMAP, Wilhelm, Holzapfel H., Stiles, Michael E. Enterococci at the crossroads of food safety? 1999;47:1–24.

32. Pereira CA, Marra AR, Camargo LF, Pignatari AC, Sukiennik T, Behar PR, Medeiros EA, Ribeiro J, Girao E, et al. Nosocomial bloodstream infections in brazilian pediatric patients: microbiology, epidemiology, and clinical features. PLOS One. 2013;8:e68144.

33. Marteau P, Cellier C. Immunological Effects of Biotherapeutic Agents. From:Biotherapeutic Agents and Infectious Diseases: Humana Press; 1999. p. 121-44.

34. Carey CM, Kostrzynska M. Lactic acid bacteria and bifidobacteria attenuate the proinflammatory response in intestinal epithelial cells induced by Salmonella enterica serovar Typhimurium. Can J Microbiol. 2013;59:9-17.

35. Morson BC. Carcinoma Arising from Areas of Intestinal Metaplasia in the Gastric Mucosa. British Journal of Cancer. 1955;9:377-85.

36. Mannick EE, Bravo LE, Zarama G, Realpe JL, Zhang XJ, Ruiz B, Fontham ET, Mera R, Miller MJ, Correa P. Inducible nitric oxide synthase, nitrotyrosine, and apoptosis in Helicobacter pylori gastritis: effect of antibiotics and antioxidants. Cancer Res. 1996;56:3238-43.

37. Bartsch H, Nair J. Chronic inflammation and oxidative stress in the genesis and perpetuation of cancer: role of lipid peroxidation, DNA damage, and repair. Langenbecks Arch Surg. 2006;391:499-510.

Page 123: To my parents for their unyielding support and encouragement

123

38. Aschengrau A, Seage GRI. Experimental Studies. Essentials of Epidemiology in Public Health. 2nd ed. United States of America: Jones and Bartlett Publishers; 2008. p. 169-200.

39. Dimenas E, Glise H, Hallerback B, Hernqvist H, Svedlund J, Wiklund I. Quality of life in patients with upper gastrointestinal symptoms. An improved evaluation of treatment regimens? Scand J Gastroenterol. 1993;28:681-7.

40. Svedlund JS, I. Dotevall, G. GSRS-a clinical rating scale for gastrointestinal symptoms in patients with irritable bowel syndrome and peptic ulcer disease. Digestive Diseases and Sciences. 1988;33:129-34.

41. Kulich KR, Madisch A, Pacini F, Pique JM, Regula J, Van Rensburg CJ, Ujszaszy L, Carlsson J, Halling K, Wiklund IK. Reliability and validity of the Gastrointestinal Symptom Rating Scale (GSRS) and Quality of Life in Reflux and Dyspepsia (QOLRAD) questionnaire in dyspepsia: a six-country study. Health Qual Life Outcomes. 2008;6:12.

42. Revicki DA, Wood M, Wiklund I, Crawley J. Reliability and validity of the gastrointestinal symptom rating scale in patients with gastroesophageal reflux disease. Quality of Life Research. 1998;7:75.

43. Rush DR, Stelmach WJ, Young TL, Kirchdoerfer LJ, Scott-Lennox J, Holverson HE, Sabesin SM, Nicholas TA. Clinical effectiveness and quality of life with ranitidine vs placebo in gastroesophageal reflux disease patients: a clinical experience network (CEN) study. J Fam Pract. 1995;41:126-36.

44. Chal KL, Stacey JH, Sacks GE. The effect of ranitidine on symptom relief and quality of life of patients with gastro-oesophageal reflux disease. Br J Clin Pract. 1995;49:73-7.

45. Helmerhorst HJ, Brage S, Warren J, Besson H, Ekelund U. A systematic review of reliability and objective criterion-related validity of physical activity questionnaires. International Journal of Behavioral Nutrition and Physical Activity. 2012;9:103.

46. Bull FC, Maslin TS, Armstrong T. Global physical activity questionnaire (GPAQ): nine country reliability and validity study. J Phys Act Health. 2009 Nov;6:790-804.

47. Tompkins TA, Hagen KE, Wallace TD, Fillion-Forte V. Safety evaluation of two bacterial strains used in Asian probiotic products. Canadian Journal of Microbiology. 2008;54:391-400.

48. Casula G, Cutting SM. Bacillus Probiotics: Spore Germination in the Gastrointestinal Tract. 2002 2002-05-01.

49. Tompkins TA, Xu X, Ahmarani J. A comprehensive review of post-market clinical studies performed in adults with an Asian probiotic formulation. Benef Microbes. 2010;1:93-106.

Page 124: To my parents for their unyielding support and encouragement

124

50. Khodadad A, Farahmand F, Najafi M, Shoaran M. Probiotics for the Treatment of Pediatric Helicobacter Pylori Infection: A Randomized Double Blind Clinical Trial. Iran J Pediatr. 2013;23:79-84.

51. Bekar O, Yilmaz Y, Gulten M. Kefir improves the efficacy and tolerability of triple therapy in eradicating Helicobacter pylori. J Med Food. 2011;14:344-7.

52. Sheu BS, Wu JJ, Lo CY, Wu HW, Chen JH, Lin YS, Lin MD. Impact of supplement with Lactobacillus- and Bifidobacterium-containing yogurt on triple therapy for Helicobacter pylori eradication. Aliment Pharmacol Ther. 2002;16:1669-75.

53. Sorokulova IB, Pinchuk IV, Denayrolles M, Osipova IG, Huang JM, Cutting SM, Urdaci MC. The safety of two Bacillus probiotic strains for human use. Dig Dis Sci. 2008;53:954-63.

54. Lipinski S, Till A, Sina C, Arlt A, Grasberger H, Schreiber S, Rosenstiel P. DUOX2-derived reactive oxygen species are effectors of NOD2-mediated antibacterial responses. J Cell Sci. 2009;122:3522-30.

55. Granum PE, Lund T. Bacillus cereus and its food poisoning toxins. FEMS Microbiology Letters. 1997;157:223-8.

56. Hata Y, Yamamoto M, Ohni M, Nakajima K, Nakamura Y, Takano T. A placebo-controlled study of the effect of sour milk on blood pressure in hypertensive subjects. American J of Clin Nutr. 1996;64:767-71.

57. Nicholson WL, Munakata N, Horneck G, Melosh HJ, Setlow P. Resistance of Bacillus Endospores to Extreme Terrestrial and Extraterrestrial Environments. Microbiol Mol Biol Rev. 2000;64:548-72.

58. Spinosa MR, Braccini T, Ricca E, Felice MD, Morelli L, Pozzi G, Oggioni MR. On the fate of ingested Bacillus spores. Res Microbiol. 2000;151:361–8.

59. Lysenko E, Ogura T, Cutting SM. Characterization of the ftsH gene of Bacillus subtilis. Microbiology. 1997;43:971-8.

60. Hong HA, Khaneja R, Tam NMK, Cazzato A, Tan S, Urdaci M, Brisson A, Gasbarrini A, Barnes I, Cutting SM. Bacillus subtilis isolated from the human gastrointestinal tract. Research in Microbiology. 2009;160:134-43.

61. Fakhry S, Sorrentini I., Ricca, E.,DeFelice, M., Baccigalupi, L. Characterization of spore forming Bacilli isolated from the human gastrointestinal tract. Journal of Applied Microbiology. 2008;105:2178-86.

62. Hong HA, To E, Fakhry S, Baccigalupi L, Ricca E, Cutting SM. Defining the natural habitat of Bacillus spore-formers. Res Microbiol. 2009;160:375-9.

Page 125: To my parents for their unyielding support and encouragement

125

63. Branda SS, González-Pastor JE, Ben-Yehuda S, Losick R, Kolter R. Fruiting body formation by Bacillus subtilis. Proceedings of the National Academy of Sciences. 2001;98:11621-6.

64. Inooka S, Uehara S, Kimura M. The effect of Bacillus natto on the T and B lymphocytes from spleens of feeding chickens. Poultry Science. 1986;65:1217-9.

65. Andersson H, Asp N-G, Bruce A, Roos S, Wadstrom T, Wold AE. Health effects of probiotics and prebioticsA literature review on human studies Scandinavian J of Nutr. 2001;45:58-75.

66. Hosoi T, Ametani A, Kiuchi K, Kaminogawa S. Improved growth and viability of lactobacilli in the presence of Bacillus subtilis (natto), catalase, or subtilisin. Can J Microbiol. 2011;46:892-7.

67. Tsukamoto Y, Ichise H, Kakuda H, Yamaguchi M. Intake of fermented soybean (natto) increases circulating vitamin K2 (menaquinone-7) and gamma-carboxylated osteocalcin concentration in normal individuals. J Bone Mineral Metabolism. 2000;18:216-22.

68. Lee H, Kim YH, Kim JH, Chang DK, Kim JY, Son HJ, Rhee PL, Kim JJ, Rhee JC. A feasibility study of probiotics pretreatment as a bowel preparation for colonoscopy in constipated patients. Dig Dis Sci. 2010;55:2344-51.

69. Lee H, Kim Y-H, Kim JH, Chang DK et al. A Feasibility Study of Probiotics Pretreatment as a Bowel Preparation for Colonoscopy in Constipated Patients. Digestive Diseases and Sciences. 2013;55:2344-51.

70. American College of Sports Medicine Position Stand. The recommended quantity and quality of exercise for developing and maintaining cardiorespiratory and muscular fitness, and flexibility in healthy adults. Med Sci Sports Exerc. 1998;30:975-91.

71. Cutting M. S. Bacillus probiotics. Food Microbiology. 2011;28:214-20.

72. Urdaci MC, Pinchuk I, Ricca E, Henriques AO, Cutting SM. Antimicrobial activity of Bacillus probiotics. Bacterial spore formers: probiotics and emerging applications: Horizon Bioscience; 2004. p. 171-82.

73. Huang JM, La Ragione RM, Nunez A, Cutting SM. Immunostimulatory activity of Bacillus spores. FEMS Immunol Med Microbiol. 2008;53:195-203.

74. Walker E, Nowacki AS. Understanding Equivalence and Noninferiority Testing. J Gen Intern Med. 2011;26:192-6.

75. Barker LE, Luman ET, McCauley MM, Chu SY. Assessing equivalence: an alternative to the use of difference tests for measuring disparities in vaccination coverage. Am J Epidemiol. 2002;156:1056-61.

Page 126: To my parents for their unyielding support and encouragement

126

BIOGRAPHICAL SKETCH

Abdulah Hanifi was born in Castro Valley, California. The youngest of four

children, he grew up mostly in Clovis, California, graduating from University High School

in 2007. He attended California State University Fresno and earned his B.A. in

philosophy with an emphasis in religious studies, and chemistry minor in 2011. He

graduated magna cum laude and was awarded a Dean’s Medal for outstanding

academic achievement in the College of Arts and Humanities. His involvement at the

University of California San Francisco-Fresno Department of Emergency Medicine’s

Academic Research Associate Program afforded him the valuable opportunity to work

closely with physicians and medical staff conducting clinical research on emergency

room patients.

His training in the humanities, sciences and clinical research allowed him the

opportunity to begin his M.S. in food science and human nutrition at the University of

Florida. He began his work studying the clinical outcomes on the consumption of a

novel probiotic in 2012 and upon completion of his M.S. program, Abdulah will continue

his studies to earn a Ph.D. in food science and human nutrition.